CA2628461A1 - Substituted 1h-benzimidazole-4-carboxamides are potent parp inhibitors - Google Patents

Substituted 1h-benzimidazole-4-carboxamides are potent parp inhibitors Download PDF

Info

Publication number
CA2628461A1
CA2628461A1 CA002628461A CA2628461A CA2628461A1 CA 2628461 A1 CA2628461 A1 CA 2628461A1 CA 002628461 A CA002628461 A CA 002628461A CA 2628461 A CA2628461 A CA 2628461A CA 2628461 A1 CA2628461 A1 CA 2628461A1
Authority
CA
Canada
Prior art keywords
benzimidazole
carboxamide
therapeutically acceptable
formula
group
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
CA002628461A
Other languages
French (fr)
Other versions
CA2628461C (en
Inventor
Thomas D. Penning
Sheela A. Thomas
Gui-Dong Zhu
Jianchun Gong
Vincent L. Giranda
Viraj B. Gandhi
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
AbbVie Inc
Original Assignee
Abbott Laboratories
Thomas D. Penning
Sheela A. Thomas
Gui-Dong Zhu
Jianchun Gong
Vincent L. Giranda
Viraj B. Gandhi
Abbvie Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Abbott Laboratories, Thomas D. Penning, Sheela A. Thomas, Gui-Dong Zhu, Jianchun Gong, Vincent L. Giranda, Viraj B. Gandhi, Abbvie Inc. filed Critical Abbott Laboratories
Publication of CA2628461A1 publication Critical patent/CA2628461A1/en
Application granted granted Critical
Publication of CA2628461C publication Critical patent/CA2628461C/en
Expired - Fee Related legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D235/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings
    • C07D235/02Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings condensed with carbocyclic rings or ring systems
    • C07D235/04Benzimidazoles; Hydrogenated benzimidazoles
    • C07D235/18Benzimidazoles; Hydrogenated benzimidazoles with aryl radicals directly attached in position 2
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/06Antigout agents, e.g. antihyperuricemic or uricosuric agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/02Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/02Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings
    • C07D409/04Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond

Abstract

Compounds of Formula (I) inhibit the PARP enzyme and are useful for treating a disease or a disorder associated with PARP. Also disclosed are pharmaceutical compositions comprising compounds of Formula (I), methods of treatment comprising compounds of Formula (I), and methods of inhibiting the PARP enzyme comprising compounds of Formula (I).

Description

INHIBITORS
Technical Field The present invention relates to 1H-benzimidazole-4-carboxamides, their preparation, and their use as inhibitors of the enzyme poly(ADP-ribose)polymerase for the preparation of drugs.

Back round Poly(ADP-ribose)polymerase (PARP) or poly(ADP-ribose)synthase (PARS) has an essential role in facilitating DNA repair, controlling RNA transcription, mediating cell death, and regulating immune response. These actions make PARP inhibitors targets for a broad spectrum of disorders. PARP inhibitors have demonstrated efficacy in numerous models of disease, particularly in models of ischemia reperfusion injury, inflammatory disease, degenerative diseases, protection from adverse effects of cytoxic compounds, and the potentiation of cytotoxic cancer therapy. PARP has also been indicated in retroviral infection and thus inhibitors may have use in antiretroviral therapy. PARP inhibitors have been efficacious in preventing ischemia reperfusion injury in models of myocardial infarction, stroke, other neural trauma, organ transplantation, as well as reperfusion of the eye, kidney, gut and skeletal muscle. Inhibitors have been efficacious in inflammatory diseases such as arthritis, gout, inflammatory bowel disease, CNS inflammation such as MS and allergic encephalitis, sepsis, septic shock, hemmorhagic shock, pulmonary fibrosis, and uveitis.
PARP inhibitors have also shown benefit in several models of degenerative disease including diabetes (as well as complications) and Parkinsons disease. PARP inhibitors can ameliorate the liver toxicity following acetominophen overdose, cardiac and kidney toxicities from doxorubicin and platinum based antineoplastic agents, as well as skin damage secondary to sulfur mustards. In various cancer models, PARP inhibitors have been shown to potentiate radiation and chemotherapy by increasing cell death of cancer cells, limiting tumor growth, decreasing metastasis, and prolonging the survival of tumor-bearing animals.
Summary of the Invention In one embodiment, the present invention provides compounds of Formula (I) R, 1 N
\~--X'-W
R N
(I), or a therapeutically acceptable salt, prodrug, or salt of a prodrug thereof, wherein Rl, R2, and R3 are independently selected from the group consisting of hydrogen, alkenyl, alkoxy, alkoxycarbonyl, alkyl, alkynyl, cyano, haloalkoxy, haloalkyl, halogen, hydroxy, hydroxyalkyl, nitro, NRARB, and (NRARB)carbonyl;
' XI is selected from the group consisting of aryl and heteroaryl, wherein XI
is optionally substituted with 1, 2, 3, or 4 substituents selected from the group consisting of alkyl, cyano, halogen, and haloalkyl;
W is X2-Y-NR6R7 or Y-NR6R7;
X2 is selected from the group consisting of aryl and heteroaryl, wherein X2 is optionally substituted with 1, 2, 3, or 4 substituents selected from the group consisting of alkyl, cyano, halogen, and haloalkyl;
Y is alkylenyl;
R6 is selected from the group consisting of hydrogen, alkenyl, alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, alkyl, alkynyl, aryl, arylalkyl, cycloalkyl, cycloalkylalkyl, heterocycle, heterocyclealkyl, heteroaryl, heteroarylalkyl, hydroxyalkyl, (NRCRD)alkyl, (NRcRD)carbonyl, (NRcRD)carbonylalkyl, and (NRcRD)sulfonyl;
R7 is selected from the group consisting of cycloalkyl and cycloalkyl fused to phenyl, wherein R7 may be optionally substituted with 1, 2, or 3 substituents selected from the group consisting of alkenyl, alkoxy, alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, alkyl, alkylcarbonyl, alkylcarbonylalkyl, alkynyl, carboxy, cycloalkyl, cycloalkylalkyl, cyano, haloalkoxy, haloalkyl, halogen, hydroxy, hydroxyalkyl, NRcRn, (NRCRD)alkyl, (NRcRD)carbonyl, (NRcRD)carbonylalkyl, (NRCRD)sulfonyl, and oxo; and RA, RB, Rc, and RD are independently selected from the group consisting of hydrogen, alkyl, and alkycarbonyl.

Detailed Description of the Invention In another embodiment, the present invention provides compounds of Formula (I) R, N
I N}-X1-W
R N

(I), or a therapeutically acceptable salt, prodrug, or salt of a prodrug thereof, wherein Ri, R2, and R3 are independently selected from the group consisting of hydrogen and halogen;
X1 is selected from the group consisting of aryl and heteroaryl, wherein X, is optionally substituted 'with halogen;
W is X2-Y-NR6R7 or Y-NR6R7;
X2 is aryl;

Y is alkylenyl; R6 is selected from the group consisting of hydrogen and alkyl; and R7 is selected from the group consisting of cycloalkyl and cycloalkyl fused to phenyl.
In another embodiment, the present invention provides a pharmaceutical composition comprising a compound of Formula (I), wherein Rt, R2, and R3 are hydrogen.
In another embodiment, the present invention provides a pharmaceutical composition comprising a compound of Formula (I), wherein W is Y-NR6R7.
In another embodiment, the present invention provides a pharmaceutical composition comprising a compound of Formula (I), wherein R7 is cycloalkyl.
In another embodiment, the present invention provides a pharmaceutical-composition comprising a compound of Formula (I), wherein R6 is hydrogen; and R7 is cycloalkyl.
In another embodiment, the present invention provides a pharmaceutical composition comprising a compound of Formula (I), wherein Xl is thiophenyl.
In another embodiment, the present invention provides a pharmaceutical composition comprising a compound of Formula (I), wherein X, is thiazolyl.
In another embodiment, the present invention provides a pharmaceutical composition comprising a compound of Formula (I), wherein Xt is phenyl and X2 is phenyl.
In another embodiment, the present invention provides a pharmaceutical composition comprising a compound of Formula (I), wherein XI is phenyl and W is Y-NR6R,7 In another embodiment, the present invention provides a pharmaceutical composition comprising a compound of Formula (I), wherein R2 is halogen.
In another embodiment, the present invention provides a pharmaceutical composition cornprising a compound of Formula (I), or a therapeutically acceptable salt thereof, in combination with a therapeutically acceptable carrier.
In another embodiment, the present invention provides a method of inhibiting PARP
in a mammal in recognized need of such treatment comprising administering to the mammal a therapeutically acceptable amount of a compound of Formula (I) or a therapeutically acceptable salt thereof.
In another embodiment, the present invention provides a method of treating cancer in a mammal in recognized need of such treatment comprising administering to the mammal a therapeutically acceptable amount of a compound of Formula (I) or a therapeutically acceptable salt thereof.
In another embodiment, the present invention provides a method for decreasing tumor volume in a mammal in recognized need of such treatment comprising administering to the mammal a therapeutically acceptable amount of a compound of Formula (I) or a therapeutically acceptable salt thereof.
In another embodiment, the present invention provides a method of treating leukemia, colon cancer, glioblastomas, lymphomas, melanomas, carcinomas of the breast, or cervical carcinomas in a mammal in recognized need of such treatment comprising administering to the mammal a therapeutically acceptable amount of a compound of Formula (I) or a therapeutically acceptable salt thereof.
In another embodiment, the present invention provides a method of potentiation of cytotoxic cancer therapy in a mammal in recognized need of such treatment comprising administering to the mammal a therapeutically acceptable amount of a compound of Formula (I) or a therapeutically acceptable salt thereof.
In another embodiment, the present invention provides a method of potentiation of radiation therapy in a mammal in recognized need of such treatment comprising administering to the mammal a therapeutically acceptable amount of a compound of Formula (I) or a therapeutically acceptable salt thereof.
In another embodiment, the present invention provides a method of treating ischemia reperfusion injury associated with, but not limited to, myocardial infarction, stroke, other neural trauma, and organ transplantation, in a mammal in recognized need of such treatment comprising administering to the mammal a therapeutically acceptable amount of a compound of Formula (I) or a therapeutically acceptable salt thereof.
In another embodiment, the present invention provides a method of reperfusion including, but not limited to, reperfusion of the eye, kidney, gut and skeletal muscle, in a mammal in recognized need of such treatment comprising administering to the mammal a therapeutically acceptable amount of a compound of Formula (I) or a therapeutically acceptable salt thereof.
In another embodiment, the present invention provides a method of treating inflammatory diseases including, but not limited to, arthritis, gout, inflammatory bowel disease, CNS inflammation, multiple sclerosis, allergic encephalitis, sepsis, septic shock, hemmorhagic shock, pulmonary fibrosis, and uveitis in a mammal in recognized need of such treatment comprising administering to the mammal a therapeutically acceptable amount of a compound of Formula (I) or a therapeutically acceptable salt thereof.
In another embodiment, the present invention provides a method of treating immunological diseases or disorders such as rheumatoid arthritis and septic shock in a mammal in recognized need of such treatment comprising administering to the mammal a therapeutically acceptable amount of a compound of Formula (I) or a therapeutically acceptable salt thereof.
In another embodiment, the present invention provides a method of treating degenerative disease including, but not limited to, diabetes and Parkinsons disease, in a mammal in recognized need of such treatment comprising administering to the mammal a therapeutically acceptable amount of a compound of Formula (I) or a therapeutically acceptable salt thereof.
In another embodiment, the present invention provides a method of treating hypoglycemia in a mammal in recognized need of such treatment comprising administering to the mammal a therapeutically acceptable amount of a compound of Formula (I) or a therapeutically acceptable salt thereof.
In another embodiment, the present invention provides a method of treating retroviral infection in a mammal in recognized need of such treatment comp'rising administering to the mammal a therapeutically acceptable amount of a compound of Formula (I) or a therapeutically acceptable salt thereof.

In another embodiment, the present invention provides a method of treating liver toxicity following acetominophen overdose in a mammal in recognized need of such treatment comprising administering to the mammal a therapeutically acceptable amount of a compound of Formula (I) or a therapeutically acceptable salt thereof.
In another embodiment, the present invention provides a method of treating cardiac and kidney toxicities from doxorubicin and platinum based antineoplastic agents in a mammal in recognized need of such treatment comprising administering to the mammal a therapeutically acceptable amount of a compound of Formula (I) or a therapeutically acceptable salt thereof.
In another embodiment, the present invention provides a method of treating skin damage secondary to sulfur mustards in a mammal in recognized need of such treatment comprising administering to the mammal a therapeutically acceptable amount of a compound of Formula (I) or a therapeutically acceptable salt thereof.
In another embodiment, the present invention provides a use of a compound of Formula (I), or a therapeutically acceptable salt thereof, to prepare a medicament for inhibiting the P.ARP enzyme in a mammal in recognized need of such treatment.
In another embodiment, the present invention provides a use of a compound of Formula (I), or a therapeutically acceptable salt thereof, to prepare a medicament for inhibiting tumor growth in a mammal in recognized need of such treatment.
In another embodiment, the present invention provides a use of a compound of Formula (I), or a therapeutically acceptable salt thereof, to prepare a medicament for treating cancer in a mammal in recognized need of such treatment.
In another embodiment, the present invention provides a use of a compound of Formula (I), or a therapeutically acceptable salt thereof, to prepare a medicament for treating leukemia, colon cancer, glioblastomas, lymphomas, melanomas, carcinomas of the breast, or cervical carcinomas in a mammal in a mammal in recognized need of such treatment.
In another embodiment, the present invention provides a use of a compound of Formula (I), or a therapeutically acceptable salt thereof, to prepare a medicament for potentiation of cytotoxic cancer therapy in a mammal in recognized need of such treatment comprising administering to the mammal a therapeutically acceptable amount of a compound of Formula (1) or a therapeutically acceptable salt thereof.

In another embodiment, the present invention provides a use of a compound of Forrnula (I), or a therapeutically acceptable salt thereof, to prepare a medicament for potentiation of radiation in a mammal in recognized need of such treatment comprising administering to the mammal a therapeutically acceptable amount of a compound of Formula (I) or a therapeutically acceptable salt thereof.
In another embodiment, the present invention provides a use of a compound of Formula (I), or a therapeutically acceptable salt thereof, to prepare a medicament for treating ischemia reperfusion injury associated with, but not limited to, myocardial infarction, stroke, other neural trauma, and organ transplantation, in a mammal in recognized need of such treatment comprising administering to the mammal a therapeutically acceptable amount of a compound of Formula (I). or a therapeutically acceptable salt thereof.
In another embodiment, the present invention provides a use of a compound of Formula (I), or a therapeutically acceptable salt thereof, to prepare a medicament for treating reperfusion including, but not limited to, reperfusion of the eye, kidney, gut and skeletal muscle, in a mammal in recognized need of such treatment comprising administering to the mammal a therapeutically acceptable amount of a compound of Formula (I) or a therapeutically acceptable salt thereof.
In another embodiment, the present invention provides a use of a compound of Formula (I), or a therapeutically acceptable salt thereof, to prepare a medicament for treating inflammatory diseases including, but not limited to, arthritis, gout, inflammatory bowel disease, CNS inflammation, multiple sclerosis, allergic encephalitis, sepsis, septic shock, hemmorhagic shock, pulmonary fibrosis, and uveitis in a mammal in recognized need of such treatment comprising administering to the mammal a therapeutically acceptable amount of a compound of Formula (1) or a therapeutically acceptable salt thereof.
In another embodiment, the present invention provides a use of a compound of Formula (I), or a therapeutically acceptable salt thereof, to prepare a medicament for treating immunological diseases or disorders such as rheumatoid arthritis and septic shock in a mammal in recognized need of such treatment comprising administering to the mammal a therapeutically acceptable amount of a compound of Formula (I) or a therapeutically acceptable salt thereof.
In another embodiment, the present invention provides a use of a compound of Formula (I), or a therapeutically acceptable salt thereof, to prepare a medicament for treating degenerative disease including, but not limited to, diabetes and Parkinsons disease, in a mammal in recognized need of such treatment comprising administering to the mammal a therapeutically acceptable amount of a compound of Formula (I) or a therapeutically acceptable salt thereof.
In another embodiment, the present invention provides a use of a compound of Formula (I), or a therapeutically acceptable salt thereof, to prepare a medicament for treating hypoglycemia in a mammal in recognized need of such treatnient comprising administering to the mammal a therapeutically acceptable amount of a compound of Formula (I) or a therapeutically acceptable salt thereof.
In another embodiment, the present invention provides a use of a compound of Formula. (I), or a therapeutically acceptable salt thereof, to prepare a medicament for treating retroviral infection in a mammal in recognized need of such treatment comprising administering to the mammal a therapeutically acceptable amount of a compound of Formula (I) or a therapeutically acceptable salt thereof.
In another embodiment, the present invention provides a use of a compound of Formula (I), or a therapeutically acceptable salt thereof, to prepare a medicament for treating liver toxicity following acetaminophen overdose in a mammal in recognized need of such treatment comprising administering to the mammal a therapeutically acceptable amount of a compound ofFormula (I) or a therapeutically acceptable salt thereof_ In another embodiment, the present invention provides a use of a compound of Fonnula (I), or a therapeutically acceptable salt thereof, to prepare a medicament for treating cardiac and kidney toxicities from doxorubicin and platinum based antineoplastic agents in a mammal in recognized need of such treatment comprising administering to the mammal a therapeutically acceptable amount of a compound of Formula (1) or a therapeutically acceptable salt thereof.
In another embodiment, the present invention provides a use of a compound of Formula (I), or a therapeutically acceptable salt thereof, to prepare a medicament for treating skin damage secondary to sulfur mustards in a mammal in recognized need of such treatment comprising administering to the mammal a therapeutically acceptable amount of a compound of Formula (I) or a therapeutically acceptable salt thereof.

Definitions As used throughout this specification and the appended claims, the following terms have the following meanings:
The term "alkenyl" as used herein, means a straight or branched chain hydrocarbon containing from 2 to 10 carbons and containing at least one carbon-carbon double bond formed by the removal of two hydrogens. Representative examples of alkenyl include, but are not limited to, ethenyl, 2-propenyl, 2-methyl-2-propenyl, 3-butenyl, 4-pentenyl, 5-hexenyl, 2-heptenyl, 2-methyl-l-heptenyl, and 3-decenyl.
The term "alkoxy" as used herein, means an alkyl group, as defined herein, appended to the parent molecular moiety through an oxygen atom. Representative examples of alkoxy include, but are not limited to, methoxy, ethoxy, propoxy, 2-propoxy, butoxy, tert-butoxy, pentyloxy, and hexyloxy.
The term "alkoxyalkyl" as used herein, means at least one alkoxy group, as defined herein, appended to the parent molecular moiety through an alkyl group, as defined herein.
Representative examples of alkoxyalkyl include, but are not limited to, tert-butoxymethyl, 2-ethoxyethyl, 2-methoxyethyl, and methoxymethyl.
The term "alkoxycarbonyl" as used herein, means an alkoxy group, as defined herein, appended to the parent molecular moiety through a carbonyl group, as defined herein.
Representative examples of alkoxycarbonyl include, but are not limited to, methoxycarbonyl, ethoxycarbonyl, and tert-butoxycarbonyl.
The term "alkoxycarbonylalkyl" as used herein, means an alkoxycarbonyl group, as defined herein, appended to the parent molecular moiety through an alkyl group, as defined herein.
The term "alkyl" as used herein, means a straight or branched chain hydrocarbon containing from 1 to 10 carbon atoms. Representative examples of alkyl include, but are not limited to, methyl, ethyl, n-propyl, iso-propyl, n-butyl, sec-butyl, iso-butyl, tert-butyl, n-pentyl, isopentyl, neopentyl, n-hexyl, 3-methylhexyl, 2,2-dimethylpentyl, 2,3-dimethylpentyl, n-heptyl, n-octyl, n-nonyl, and n-decyl.
The term "alkylcarbonyl" as used herein, means an alkyl group, as defined herein, appended to the parent molecular moiety through a carbonyl group, as defined herein.
Representative examples of alkyicarbonyl include, but are not limited to, acetyl, 1-oxopropyl, 2,2-dimethyl-l-oxopropyl, 1-oxobutyl, and 1-oxopentyl.

The term "alkyl.carbonyloxy" as used herein, means an alkylcarbonyl group, as defined herein, appended to the parent molecular moiety through an oxygen atom.
Representative examples of alkylcarbonyloxy include, but are not limited to, acetyloxy, ethylcarbonyloxy, and tert-butylcarbonyloxy.
The term "alkylenyl" as used herein, means a divalent group derived from a straight or branched chain hydrocarbon of from 1 to 6 carbon atoms. Representative examples include, but are not limited to, -CH2-, -CH(CH3)-, -C(CH3)2-, -CH2CH2-, -CHaCH2CHZCH2CH2- ,-CH2CHaCH2-, -CH2CH(CH3)CH2-.
The term "alkylthio" as used herein, means an alkyl group, as defined herein, appended to the parent molecular moiety through a sulfur atom. Representative examples of alkylthio include, but are not limited, methylthio, ethylthio, tert-butylthio, and hexylthio.
The term "alkylthioalkyl" as used herein, means an alkylthio group, as defined herein, appended to the parent molecular moiety through an alkyl group, as defined herein.
Representative examples of alkylthioalkyl include, but are not limited, methylthiomethyl and 2-(ethylthio)ethyl.
The ten n "alkynyl" as used herein, means a straight or branched chain hydrocarbon group containing from 2 to 10 carbon atoms and containing at least one carbon-carbon triple bond. Representative examples of alkynyl include, but are not limited, to acetylenyl, 1-propynyl, 2-propynyl, 3-butynyl, 2-pentynyl, and 1-butynyl.
The term "aryl," as used herein, means a phenyl group or a naphthyl group.
The aryl groups of the present invention can be optionally substituted with one, two, three, four, or five substituents independently selected from the group consisting of alkenyl, alkoxy, alkoxyalkyl, alkoxycarbonyl, alkyl, alkylcarbonyl, alkylcarbonyloxy, alkylthio, alkylthioalkyl, alkynyl, carboxy, cyano, formyl, haloalkoxy, haloalkyl, halogen, hydroxy, hydroxyalkyl, mercapto, nitro, -NRERF, -and (NRERF)carbonyl.
The term "arylalkyl" as used herein, means an aryl group, as defined herein, appended to the parent molecular moiety through an alkyl group, as defined herein.
Representative examples of arylalkyl include, but are not limited to, benzyl, 2-phenylethyl, 3-phenylpropyl, 1-methyl-3-phenylpropyl, and 2-naphth-2-ylethyl.
The tenn "carbonyl" as used herein, means a -C(O)- group.
The term "carboxy" as used herein, means a-CO2H group.
The term "cyano" as used herein, means a -CN group.

The term "cycloalkyl" as used herein, means a saturated cyclic hydrocarbon group containing from 3 to 8 carbons, examples of cycloalkyl include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl.
The cycloalkyl groups of the present invention are optionally substituted with 1, 2, 3, or 4 substituents selected from alkenyl, alkoxy, alkoxyalkyl, alkoxycarbonyl, alkyl, alkylcarbonyl, alkylcarbonyloxy, alkylthio, alkylthioalkyl, alkynyl, carboxy, cyano, formyl, haloalkoxy, haloalkyl, halogen, hydroxy, hydroxyalkyl, mercapto, oxo, -NRERF, and (NRERF)carbonyl.
The term "cycloalkylalkyl" as used herein, means a cycloalkyl group, as defined herein, appended to the parent molecular moiety through an alkyl group, as defined herein.
Representative examples of cycloalkylalkyl include, but are not limited to, cyclopropylmethyl, 2-cyclobutylethyl, cyclopentylmethyl, cyclohexylmethyl, and 4-cycloheptylbutyl.
The term "formyl" as used herein, means a -C(O)H group.
The term "halo" or "halogen" as used herein, means -Cl, -Br, -I or -F.
The term "haloalkoxy" as used herein, means at least one halogen, as defined herein, appended to the parent molecular moiety through an alkoxy group, as defined herein.
Representative examples of haloalkoxy include, but are not limited to, chloromethoxy, 2-fluoroethoxy, trifluoromethoxy, and pentafluoroethoxy.
The term "haloalkyl" as used herein, means at least one halogen, as defined herein, appended to the parent molecular moiety through an alkyl group, as defined herein.
Representative examples of haloalkyl include, but are not limited to, chloromethyl, 2-fluoroethyl, trifluoromethyl, pentafluoroethyl, and 2-chloro-3-fluoropentyl.
The term "heteroaryl," as used herein, means a monocyclic heteroaryl ring or a bicyclic heteroaryl ring. The monocyclic heteroaryl ring is a 5 or 6 membered ring. The 5 membered ring has two double bonds and contains one, two, three or four heteroatoms independently selected from the group consisting of N, 0, and S. The 6 membered ring has three double bonds and contains one, two, three or four heteroatoms independently selected from the group consisting of N, 0, and S. The bicyclic heteroaryl ring consists of the 5 or 6 membered heteroaryl ring fused to a phenyl group or the 5 or 6 membered heteroaryl ring is fused to another 5 or 6 membered heteroaryl ring. Nitrogen heteroatoms contained within the heteroaryl may be optionally oxidized to the N-oxide. The heteroaryl is connected to the parent molecular moiety through any carbon atom contained within the heteroaryl while maintaining proper valence. Representative examples of heteroaryl include, but are not limited to, benzothienyl, benzoxadiazolyl, cinnolinyl, furopyridinyl, furyl, imidazolyl, indazolyl, indolyl, isoxazolyl, isoquinolinyl, isothiazolyl, naphthyridinyl, oxadiazolyl, oxazolyl, pyridinyl, pyridazinyl, pyrimidinyl, pyrazinyl, pyrazolyl, pyrrolyl, pyridinium N-oxide, quinolinyl, tetrazolyl, thiadiazolyl, thiazolyl, thienopyridinyl, thienyl, triazolyl, and triazinyl.
The heteroaryl groups of the present invention are substituted with 0, 1, 2, 3, or 4 substituents independently selected from alkenyl, alkoxy, alkoxyalkyl, alkoxycarbonyl, alkyl, alkylcarbonyl, alkylcarbonyloxy, alkylthio, alkylthioalkyl, alkynyl, carboxy, cyano, formyl, haloalkoxy, haloalkyl, halogen, hydroxy, hydroxyalkyl, mercapto, nitro, -NRFRF, and (NRERF)carbonyl.
The term "heteroarylalkyl" as used herein, means a heteroaryl, as defined herein, appended to the parent molecular moiety through an alkyl group, as defined herein.
Representative examples of heteroarylalkyl include, but are not limted to, pyridinymethyl.
The terin "heterocycle" or "heterocyclic" as used herein, means a monocyclic or bicyclic heterocyclic ring. The monocyclic heterocyclic ring consists of a 3, 4, 5, 6, 7, or 8 membered ring containing at least one heteroatom independently selected from 0, N, and S.
The 3 or 4 membered ring contains 1 heteroatom selected from the group consisting of 0, N
and S. The 5 membered ring contains zero or one double bond and one, two or three heteroatoms selected from the group consisting of 0, N and S. The 6 or 7 membered ring contains zero, one or two double bonds and one, two or three heteroatoms selected from the group consisting of 0, N and S. The bicyclic heterocyclic ring consists of a monocyclic heterocyclic ring fused to a cycloalkyl group or the monocyclic heterocyclic ring fused to a phenyl group or the monocyclic heterocyclic ring fused to another monocyclic heterocyclic ring. The heterocycle is connected to the parent molecular moiety through any carbon or nitrogen atom contained within the heterocycle while maintaining proper valence.
Representative examples of heterocycle include, but are not limited to, azetidinyl, azepanyl, aziridinyl, diazepanyl, 1,3-dioxanyl, 1,3-dioxolanyl, 1,3-dithiolanyl, 1,3-dithianyl, imidazolinyl, imidazolidinyl, isothiazolinyl, isothiazolidinyl, isoxazolinyl, isoxazolidinyl, morpholinyl, oxadiazolinyl, oxadiazolidinyl, oxazolinyl, oxazolidinyl, piperazinyl, piperidinyl, pyrazolinyl, pyrazolidinyl, pyrrolinyl, pyrrolidinyl, tetrahydrofuranyl, tetrahydropyranyl, tetrahydrothienyl, thiadiazolinyl, thiadiazolidinyl, thiazolinyl, thiazolidinyl, thiomorpholinyl, 1,1-dioxidothiomorpholinyl (thiomorpholine sulfone), thiopyranyl, and trithianyl.
The heterocycles of this invention are substituted with 0, 1, 2,or 3 substituents independently selected from alkenyl, alkoxy, alkoxyalkyl, alkoxycarbonyl, alkyl, alkylcarbonyl, alkylcarbonyloxy, alkylthio, alkylthioalkyl, alkynyl, carboxy, cyano, formyl, haloalkoxy, haloalkyl, halogen, hydroxy, hydroxyalkyl, mercapto, nitro, -NRERF, and (NRERF)carbonyl.
The term "heterocyclealkyl" as used herein, means a heterocycle, as defined herein, appended to the parent molecular moiety through an alkyl group, as defined herein.
The term "hydroxy" as used herein, means an -OH group.
The term "hydroxyalkyl" as used herein, means at least one hydroxy group, as defined herein, is appended to the parent molecular moiety through an alkyl group, as defined herein.
Representative examples of hydroxyalkyl include, but are not limited to, hydroxymethyl, 2-hydroxyethyl, 3-hydroxypropyl, 2,3-dihydroxypentyl, and 2-ethyl-4-hydroxyheptyl.
The term "mercapto" as used herein, means a -SH group.
The ternl "nitro" as used herein, means a-NO2 group.
The term "nonaromatic" as used herein, means that a 4 membered nonaromatic ring contains zero double bonds, a 5 membered nonaromatic ring contains zero or one double bond, a 6, 7, or 8 membered nonaromatic ring contains zero, one, or two double bonds.
The term "NRARB" as used herein, means two groups, RA and RB, which are appended to the parent molecular moiety through a nitrogen atom. RA and RB are each independently hydrogen, alkyl, and alkylcarbonyl. Representative examples ofNRARB include, but are not limited to, amino, methylamino, acetylamino, and acetylmethylamino.
The term "(NRARB)carbonyl" as used herein, means a NRARB group, as defined herein, appended to the parent molecular moiety through a carbonyl group, as defined herein.
Representative examples of (NRARB)carbonyl include, but are not limited to, arninocarbonyl, (methylamino)carbonyl, (dimethylamino)carbonyl, and (ethylmethylamino)carbonyl.
The term "NRCRD" as used herein, means two groups, Rc and RD, which are appended to the parent molecular moiety through a nitrogen atom. RC and RD are each independently hydrogen, alkyl, and alkylcarbonyl. Representative examples of NRCRD include, but are not limited to, amino, methylamino, acetylamino, and acetylmethylamino.

The term "(NRcRD)carbonyl" as used herein, means a NRcRD group, as defined herein, appended to the parent molecular moiety through a carbonyl group, as defined herein.
Representative examples of (NRcRD)carbonyl include, but are not limited to, aminocarbonyl, (methylamino)carbonyl, (dimethylamino)carbonyl, and (ethylmethylamino)carbonyl.
The term "(NRcRo)carbonylalkyi" as used herein, means a(NRcRD)carbonyl group, as defined herein, appended to the parent molecular moiety through an alkyl group, as defined herein.
The term "(NRcRn)sulfonyl" as used herein, means a NRcRn group, as defined herein, appended to the parent molecular moiety through a sulfonyl group, as defined herein.
Representative examples of (NRcRp)sulfonyl include, but are not limited to, aminosulfonyl, (methylamino)sulfonyl, (dimethylamino)sulfonyl, and (ethylmethylamino)sulfonyl.
The term "NR.ERF" as used herein, means two groups, RE and RF, which are appended to the parent molecular moiety through a nitrogen atom. RE and RF are each independently hydrogen, alkyl, and alkylcarbonyl. Representative examples of NRERF include, but are not limited to, amino, methylamino, acetylamino, and acetylmethylamino.
The term "(NRERF)carbonyl" as used herein, means a NRERF group, as defined herein, appended to the parent molecular moiety through a carbonyl group, as defined herein.
Representative examples of (NRE2.F)carbonyl include, but are not limited to, aminocarbonyl, (methylamino)carbonyl, (dimethylamino)carbonyl, and (ethylmethylamino)carbonyl.
The tenn "oxo" as used herein, means a =0 moiety.
Compounds of the present invention can exist as stereoisomers, wherein asymmetric or chiral centers are present. Stereoisomers are designated (R) or (S) depending on the configuration of substituents around the chiral carbon atom. The terms (R) and (S) used herein are configurations as defined in IUPAC 1974 Recommendations for Section E, Fundamental Stereochemistry, Pure Appl. Chem., (1976), 45: 13-30, hereby incorporated by reference. The present invention contemplates various stereoisomers and mixtures thereof and are specifically included within the scope of this invention.
Stereoisomers include enantiomers, diastereomers, and mixtures of enantiomers or diastereomers.
Individual stereoisomers of compounds of the present invention may be prepared syiitheticaily from commercially available starting materials which contain asynunetric or chiral centers or by preparation of racemic mixtures followed by resolution well-known to those of ordinary skill in the art. These methods of resolution are exemplified by (1) attachment of a mixture of enantiomers to a chiral auxiliary, separation of the resulting mixture of diastereomers by recrystallization or chromatography and liberation of the optically pure product from the auxiliary or (2) direct separation of the mixture of optical enantiomers on chiral chromatographic columns.
Compounds of the present invention were named by ACD/ChemSketch version 5.06 (developed by Advanced Chemistry Development, Inc., Toronto, ON, Canada) or were given names which appeared to be consistent with ACD nomenclature.

Determination of Biological Activity Inhibition of PARP
Nicotinamide[2,5',S-3H]adenine dinucleotide and strepavidin SPA beads were purchased from Amersham Biosiences (UK) Recombinant Human Poly(ADP-Ribose) Polymerase (PARP) purified from E.coli and 6-Biotin-17-NAD+ , were purchase from Trevigen, Gaithersburg, MD. NAD+, Histone, aminobenzamide, 3-amino benzamide and Calf Thymus DNA (dcDNA) were purchased from Sigma, St. Louis, MO. Stem loop oligonucleotide containing MCAT sequence was obtained from Qiagen. The oligos were dissoloved to 1mM in annealing buffer containing 10mM Tris HCl pH 7.5, 1mM
EDTA, and 50mM NaCl, incubated for 5min at 95 C, and followed by annealing at 45 C for 45 minutes.
Histone H1 (95% electrophoretically pure) was purchased from Roche, Indianapolis, IN.
Biotinylated histone H1 was prepared by treating the protein with Sulfo-NHS-LC-Biotin from Pierce Rockford, IL. The biotinylation reaction was conducted by slowly and intermittently adding 3 equivalents of 10mM Sulfo-NHS-LC-Biotin to 100 M
Histone H1 in phosphate-buffered saline, pH 7.5, at 4 C with gentle vortexing over lmin followed by subsequent 4 C incubation for lhr. Streptavidin coated (FlashPlate Plus) microplates were purchased from Perkin Elmer, Boston, MA.
PARP1 assay was conducted in PARP assay buffer containing 50 mM Tris pH 8.0, 1mM DTT, 4 mM MgC12. PARP reactions contained 1.5 M [3H]-NAD+ (1.6uCi/mmol), 200 nM biotinylated histone H1, 200 nM sIDNA, and 1nM PARP enzyme. Auto reactions utilizing SPA bead-based detection were carried out in 100 l volumes in white 96 well plates. Reactions were initiated by adding 50 l of 2X NAD+ substrate mixture to 50 1 of 2X
enzyme mixture containing PARP and DNA. These reactions were terminated by the addition of 150 l of 1.5 mM benzamide (-1000-fold over its IC50). 170 l of the stopped reaction mixtures were transferred to streptavidin Flash Plates, incubated for lhr, and counted using a TopCount microplate scintillation counter. The Ki data was deterrnined from inhibition curves at various substrate concentrations and are shown in Table 1 for compounds of the present invention Table I
Inhibition of PARP (nM) 17 4.4 13.7 20.3 9.8 4 8.6 4.6 15.2 5.1 9 1.5 9 2.2 11.8 14.3 69 17.5 4.7 7.3 2.3 6.4 19 28 Cellular PARP assay:
C41 cells were treated with a compound of the present invention for 30 minutes in 96 well plate. PARP was then activated by damaging DNA with 1 mM H202 for 10 minutes.
The cells were then washed with ice-cold PBS once and fixed with pre-chilled methanol:acetone (7:3) at -20 C for 10 minutes. After air-drying, the plates were rehydrated with PBS and blocked 5% non-fat dry milk in PBS-tween (0.05%) (blocking solution) for 30 minutes at room temperature_ The cells were incubated with anti-PAR antibody 10H (1:50) in Blocking solution at 37 C for 60 minutes followed by washing with PBS-Tween20 5 times, and incubation with goat anti-mouse fluorescein 5(6)-isothiocyanate-coupled antibody (1:50) and 1 g/ml 4',6-diamidino-2-phenylindole (DAPI) in blocking solution at 37 C for 60 minutes. After washing with PBS-Tween20 5 times, the analysis was performed using an finax Fluorescence Microplate Reader (Molecular Devices, Sunnyvalle, CA), set at the excitation wavelength of 490 nm and emis'sion wavelength of 528 nrn fluorescein 5(6)-isothiocyanate (FITC) or the excitation wavelength of 355 nm and emission wavelength of 460 nm (DAPI). The PARP activity (FITC signal) was normalized with cell numbers (DAPI).
The cellular assay measures the fozmation of poly ADP-ribose by PARP within cells and demonstrates that compounds of the present invention penetrate cell membranes and inhibit PARP in intact cells. The ECsoS for representative compounds of the present invention are provided in Table 2.
Table 2 Cellular Activity ECSO (nM) 2.9 1.2 70 29 7.5 1.5 55 23 61 79 0.9 6 4.7 19.5 3.4 6.3 10.8 2 'I 9.8 13 3.7 As PARP inhibitors, the compounds of the present invention have numerous therapeutic applications related to, ischemia reperfusion injury, inflammatory diseases, degenerative diseases, protection from adverse effects of cytotoxic compounds, and potentiation of cytotoxic cancer therapy. In particular, compounds of the present invention potentiate radiation and chemotherapy by increasing cell death of cancer cells, limiting tumor growth, decreasing metastasis, and prolonging the survival of tumor-bearing mammals.
Compounds of Fomula (I) can treat leukemia, colon cancer, glioblastomas, lymphomas, melanomas, carcinomas of the breast, and cervical carcinomas.
Other therapeutic applications include, but are not limited to, retroviral infection, arthritis, gout, inflammatory bowel disease, CNS inflammation, multiple'sclerosis, allergic encephalitis, sepsis, septic shock, hemmorhagic shock, pulmonary fibrosis, uveitis, diabetes, Parkinsons disease, myocardial infarction, stroke, other neural trauma, organ transplantation, reperfusion of the eye, reperfusion of the kidney, reperfusion of the gut, reperfusion of skeletal muscle, liver toxicity following acetominophen overdose, cardiac and kidney toxicities from doxorubicin and platinum based antineoplastic agents, and skin damage secondary to sulfur mustards. (G. Chen et al. Cancer Chemo. Pharmacol. 22 (1988), 303; C.
Thiemermann et al., Proc. Natl. Acad. Sci. USA 94 (1997), 679-683 D. Weltin et al. Int. J.
Iinmunopharmacol. 17 (1995), 265- 271; H. Krciger et al. Inflammation 20 (1996), 203-215;
W. Ehrlich et al. Rheumatol. Int. 15 (1995), 171-172; C. Szabo et al., Proc.
Nati. Acad. Sci.
USA 95 (1998), 3867-3872; S. Cuzzocrea et al. Eur. J. Pharmacol. 342 (1998), 67-76; V.
Burkhart et al., Nature Medicine (1999), 5314-19).
When used in the above or other treatments, a therapeutically effective amount of one of the compounds of the present invention can be employed as a zwitterion or as a pharmaceutically acceptable salt. By a "therapeutically effective amount" of the compound of the invention is meant a sufficient amount of the compound to treat or prevent a disease or disorder ameliorated by a PARP inhibitor at a reasonable benefit/risk ratio applicable to any medical treatment. It will be understood, however, that the total daily usage of the compounds and compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment. The specific therapeutically effective dose level for any particular patient will depend upon a variety of factors including the disorder beirig treated and the severity of the disorder; activity of the specific compound employed; the specific composition employed, the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific compound employed; and like factors well known in the medical arts. For example, it is well within the skill of the art to start doses of the compound at levels lower than those required to achieve the desired therapeutic effect and to gradually increase the dosage until the desired effect is achieved.
The term "pharmaceutically acceptable salt" is meant to include those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like and are commensurate with a reasonable benefit/risk ratio. Pharmaceutically acceptable salts are well-known in the art. The salts can be prepared in situ during the final isolation and purification of the compounds of the present invention or separately by reacting the free base of a compound of the present invention with a-suitable acid. Representative acids include, but are not limited to acetatic, citric, aspartic, benzoic, benzenesulfonic, butyric, fumaric, hydrochloric, hydrobromic, hydroiodic, lactic, maleic, methanesulfonic, pamoic, pectinic, pivalic, propionic, succinic, tartaric, phosphic, glutamic, and p-toluenesulfonic. Also, the basic nitrogen-containing groups can be quaternized with such agents as lower alkyl halides such as methyl, ethyl, propyl, and butyl chlorides, bromides and iodides;
dialkyl sulfates like dimethyl, diethyl, dibutyl and diamyl sulfates; long chain halides such as decyl, lauryl, myristyl and stearyl chlorides, bromides and iodides; arylalkyl halides like benzyl and phenethyl bromides and others. Water or oil-soluble or dispersible products are thereby obtained.
A compound of the present invention may be administered as a pharmaceutical composition containing a compound of the present invention in combination with one or more pharmaceutically acceptable excipients. A pharmaceutically acceptable carrier or excipient refers to a non-toxic solid, semi-solid or liquid filler, diluent, encapsulating material or formulation auxiliary of any type. The compositions can be administered parenterally, intracisternally, intravaginally, intraperitoneally, topically (as by powders, ointments, drops or transdermal patch), rectally, or bucally. The term "parenteral" as used herein refers to modes of administration which include intravenous, intramuscular, intraperitoneal, intrastemal, subcutaneous and intraarticular injection and infusion.
Pharmaceutical compositions for parenteral injection comprise pharmaceutically-acceptable sterile aqueous or nonaqueous solutions, dispersions, suspensions or emulsions, as well as sterile powders for reconstitution into sterile injectable solutions or dispersions just prior to use. Examples of suitable aqueous and nonaqueous carriers, diluents, solvents or vehicles include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), carboxymethylcellulose and suitable mixtures thereof, vegetable oils (such as olive oil), and injectable organic esters such as ethyl oleate.
Proper fluidity may be maintained, for example, by the use of coating materials such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants.
These compositions can also contain adjuvants such as preservative, wetting agents, emulsifying agents, and dispersing agents. Prevention of the action of microorganisms may be ensured by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol sorbic acid, and the like. It may also be desirable to include isotonic agents such as sugars, sodium chloride, and the like. Prolonged absorption of the injectable pharmaceutical form may be brought about by the inclusion of agents which delay absorption, such as aluminum monostearate and gelatin.
Compounds of the present invention may also be administered in the form of liposomes. As is known in the art, liposomes are generally derived from phospholipids or other lipid substances. Liposomes are formed by mono- or multi-lamellar hydrated liquid crystals that are dispersed in an aqueous medium. Any non-toxic, physiologically-acceptable and metabolizable lipid capable of forming liposomes can be used. The present compositions in liposome form can contain, in addition to a compound of the present invention, stabilizers, preservatives, excipients, and the like. The preferred lipids are the phospholipids and the phosphatidyl cholines (lecithins), both natural and synthetic. Methods to form liposomes are known in the art. See, for example, Prescott, Ed., Methods in Cell Biology, Volume XIV, Academic Press, New York, N.Y. (1976), p. 33 et seq.
Total daily dose of the compositions of the invention to be administered to a human or other mammal host in single or divided doses may be in amounts, for example, from 0.0001 to 300 mg/kg body weight daily and more usually 1 to 300 mg/kg body weight.
The dose, from 0.000 1 to 300 mg/kg body, may be given twice a day.

Abbreviations which have been used in the descriptions of the examples that follow are: DBU for 1,8-diazabicyclo[5.4.0]undec-7-ene; DMF for N,N-dimethylformamide; DMSO
for dimethylsulfoxide; EtaO for diethyl ether; EtOAc for ethyl acetate; EtOH
for ethanol;
HPLC for high pressure liquid chromatography; LDA for lithium diisopropylamide; MeOH
for methanol; psi for pounds per square inch; TFA for trifluoroacetic acid;
THF for tetrahydrofu.ran, and TMS for trimethylsilane.
Compounds having formula I may be made by synthetic chemical processes, examples of which are shown herein below. It is meant to be understood that the order of the steps in the processes may be varied, that reagents, solvents and reaction conditions may be substituted for those specifically mentioned, and that vulnerable moieties may be protected and deprotected, as necessary.

Scheme 1 O NHZ O x RR O ::Hxj:

R3 ~-XjR9 R3 4 HO

Ri N~ ~O ~R7NH 30. 1 I N' R ~ N Xl R5 NaBH3CN R2 H Rs 2 H MeOH
Rg R3 $ 6 As shown in Scheme 1, compounds of formula 6 which are representative of compounds of formula (I), wherein X, is phenyl or thiophenyl and W is Y-NR6R7 may be prepared as outlined. Accordingly, when a mixture of compounds of formula 1 and compounds of formula 2 wherein R5 is hydrogen or alkyl, R8 and R9 are both an alkyl acetal or R8 and R9 taken together form a cyclic acetal, or Rs and R9 together form oxo, are heated in the presence of an acid or under conditions such as refluxing methanol containing Pd/C

will provide compounds of fonnula 4 (or formula 5 if starting with an unprotected ketone or aldehyde). Alternatively, compounds of formula 1 when treated with compounds of formula 3 (containing a protected or unprotected ketone or aldehyde similar to compounds of formula 2) using standard amide forming reagents, such as 1,1'-carbonyldiimidazole (CDI), will provide an intermediate amide, which when treated with an acid, such as acetic acid under heated conditions will provide compounds of formula 4 (or 5 if starting with an unprotected ketone or aldehyde). When compounds of formula 4 containing a protected ketone or aldehyde such as a ketal or acetal are treated with a catalytic amount of an acid-such as acetic, dilute hydrochloric or sulfuric acid will provide compounds of formula 5. Compounds of formula 5 when treated with amines R6R7NH in the presence of a reducing agent, such as but not limited to sodium cyanoborohydride or other conditions known to one skilled in the art that will effect a reductive aminiation, will provide compounds of formula 6 which are representative of compounds of the present invention.

Scheme 2 O NH2 Q-Xr-CHO 7 R (HO)2B-X2 R5 R1 ~ NH2 or ~ N 10 I ~-XT-Q

R2 NHZ Q- R3 H g X2=phenyl R3 1 X~-C02H 8 R5= hydrogen or alkyl Q=Br,CI,I
Xj=phenyl R
R, N _ RsR,NH R, N R
~ 7 N~XIX2 R5 NaBH3CN N Xl-X2 R
R2I H MeOH R2 H 6 As outlined in Scheme 2, compounds of formula 12 which are representative of compounds of formula (I) wherein both X, and X2 are phenyl may be prepared accordingly.
Compounds of formula 1 when treated with compounds of formula 7 or of formula 8, wherein X, is phenyl and Q is chloro, bromo or iodo, according to the conditions outlined in Scheme 1, which are used to generate benzanvdazoles, will provide compounds of formula 9.
Furthermore, compounds of formula 9 when treated with boronic acids of formula 10, wherein X2 is phenyl in the presence of a palladium catalyst such as but not limited to [1,1'-bis(diphenylphosphino)ferrocene]dichloro palladium and sodium carbonate under heated conditions will provide compounds of formula 11. Compounds of formula 11 when treated with amines of formula RbR7NH under reductive amination conditions known to one skilled in the art, or as outlined in Scheme 1, will provide compounds of formula 12.

Scheme 3 OH C-X j-(CH2)nCR5(Re)(Rs) Rti NH2 13 Ri N
or I \>-XI-{CH2)nCR5(Ra)(Rs) R2 NH2 HO2C-Xy-(CH2)nCRs(Ra)(Rs) R2 H

R8+R9 = -O(CH2)mO- or each 0-alkyl n = 1-4 Ry N~Xj (CH2)nC(O) 5~~ R,NH Rt N~Xj (CH2)nCHNR6R7 N Na8H3CN
R2 H MeOH R2 H
Rs 16 Rs 17 Similarly, compounds of formula 17, which are representative of compounds of formula (I), wherein Xt is phenyl and W is Y-NR6R7 are synthesized accordingly.
Compounds of formula 1 when treated with compounds of formula 13 or 14, wherein Rg and R9 are each -0-alkyl or together form a cyclic acetal, according to the procedures outlined in Scheme 1, will provide compounds of formula 15. The acetal group of compounds of formula 15 can be deprotected under acid-catalyzed conditions to provide compounds of formula 16. Compounds of formula 16 when treated to reductive amination conditions as outlined in Scheme 1 or 2 will provide compounds of formula 17.
The following Examples are intended as an illustration of and not a limitation upon the scope of the invention as defined in the appended clainis. The compounds of this invention can be prepared by a variety of synthetic routes.

Scheme 4 O Nt-12 O~X NHP O NHZ
' R, \ NHZ H 18 s Ri N NHP
I E / -X'- ' Rz NH2 0 or NHP Rz R3 H Rs R,oC(O)RIt R6 H R12C(O)RI3 R, N NH2 22 _ Ri I N' N' 24 -X' ~ N/ R NaBH3CN
Rz H RS MeOH NaBH CN Rz H 5 MeOH

R, N ~ ~ R~
~ Xi' \

Scheme 4 Scheme 4 shows the synthesis of compounds of formula 22 where Xl is thiazolyl and W is Y-NR6R7. Compounds of formula I may be coupled to compounds of formula 18 (containing an amine with a protecting group P) using, for example, Pd/C or sodium bisulfite, with heating, to give compounds of formula 20. Alternatively, compounds of formula 1 may be coupled to compounds of formula 19 (containing an amine with a protecting group P) using standard amide forming reagents, such as 1,1'-carbonyldiimidazole (CDn, to give the intermediate amide, which was treated with acid, such as acetic acid, with heating, to provide compounds of formula 20. Deprotection of the amine provided compounds of the formula 21. Compounds of formula 21 may under go reductive amination with a ketone or aldehyde 22 using conditions known to those of ordinary skill in the art to give compounds of formula 23. Compounds of formula 23 may optionally under go a second reductive amination with a ketone or aldehyde 24 to give compounds of forrnula 25.

The following Examples are intended as an illustration of and not a limitation upon the scope of the invention as defined in the appended claims. The compounds of this invention can be prepared by a variety of synthetic routes_ 2- {4-[1-(cyclohexylmethylamino)ethyl]phenyl} - 1H-benzimidazole-4-carboxamide 2-(4-acetylphenyl)-1 H-benzimi dazole-4-carboxamide 4-Acetylbenzoic acid (1.64 g, 10 mmol) in N, N-dimethylformamide (DMF, 10 mL) and pyridine (10 mL) was stirred at 40 C for 10 minutes. 1,1'-carbonyldiimidazole (CDI, 1.7 g, 10.5 mmol) was added and the mixture was stirred at 40 'C for 30 minutes. 2,3-diaminobenzamide dihydrochloride (synthesized as described in US 6737421, column 11, EXAMPLE 2, step (e), 2.2 g, 10 nunol) was added and the mixture was stirred at ambient temperature for 2.5 hours. Isopropyl alcohol (20 mL) was added and the mixture was stirred at ambient temperature for 20 hours. The resulting solid was filtered, washed with isopropyl alcohol and dried to give 2.1 g of a bright yellow solid. The crude material was stirred in water (30 mL) with 50% sodium hydroxide (1 mL) at ambient temperature for 7.5 hours. The solution was filtered and the solid (1.84 g) collected and stirred in refluxing acetic acid (25 mL) for 4 hours. The mixture was concentrated, stirred in dichloromethane, filtered, and dried to provide 1.78 g(64 fo for 2 steps) of the title compound. 1H NMR (DMSO-d6) S 9.27 (br, 1H), 8.40 (d, J=8.4 Hz, 2H), 8.16 (d, J=8.5 Hz, 2H), 7.89 (d, J=7.5 Hz, 1H), 7.78 (d, J=6.8 Hz, 1H), 7.78 (br, IH), 7.37 (t, J=7.8 Hz, IH), 2.66 (s, 3H).

2- {4-[ 1-(cyclohexylmethylamino) ethyl]phenyl} -1 H-benzimidazole-4-carboxamide A solution of EXAMPLE IA (0.1 g, 0.4 mmol) and cyclohexylmethylamine (0.2 mL, 1.5 mmol) in methanol (1 mL) was treated with sodium cyanoborohydride (0.05 g, 0.8 mmol) and acetic acid (0.1 mL). The mixture was stirred at 70 C for 72 hours then concentrated.
The residue was purified by HPLC on a C18 column with 0-100%
acetonitrile/water/ 0.1%
trifluoroacetic acid to provide the title compound as the trifluoroacetate salt. 1H NMR
(DMSO-d6) S 9.25 (s, 1H), 8.35 (s, 2H), 7.89 (s, 1H), 7.78 (s, 4H), 7.35 -7.40 (m, 1H), 4.76 (s, 1H), 3.35 (s, 6H), 2.82 (d, J= 4.9 Hz, 111), 2.46 (d, J=5.2 Hz, 1H), 1.69 (s, 2H), 1.66 (d, J=
6.4 Hz, 3H), 1.37 (s, 2H), 1.09 (s, 1H).

2-[4-(1-cyclobutylaminoethyl)phenyl]-1 H-benzimidazole-4-carboxamide A solution of EXAMPLE 1A (0.07 g, 0.3 mmol) and cyclobutylamine (0.34 mL, 4.8 mmol) in methanol (1 mL) was treated with sodium cyanoborohydride (0.016 g, 0.3 rnmol) and acetic acid (0.2 mL). The mixture was stirred at ambient temperature for 48 hours then concentrated. The residue was purified by chromatography on a silica gel column with 0-10% methanoUdichloromethane/0.1 % ainmonium hydroxide to provide the title compound.
'H NNIl2 (DMSfl-d6) 8 9.34 (s, 1H), 8.19 (d, J=7.7 Hz, 2H), 7.86 (d, J=7.7 Hz, 1H), 7.72 (d, J=8.0 Hz, 2H), 7.57 (d, J=7.7 Hz, 2H), 7.34 (t, J=7.7 Hz, 1 H), 3.97 (s, 1 H), 3.16 (d, J=5.2 Hz, 4H), 2.09 (s, 1H), 1.88 (s, 2H), 1.75 (s, 1H), 1.60 (s, 1H), 1.52 (s, 1H), 1.36 (s, 3H).

2- {4'-[(cyclohexylmethylamino)methyl]biphenyl-4-yl} -1 H-benzimidazole-4-carboxamide 2-(4-bromophenyl)-1 H-b enzimidazole-4-carboxamide To a mixture of 2,3-diaminobenzamide dihydrochloride (synthesized as described in US 6737421, column 11, EXAMPLE 2, step (e), 5 g, 22.3 mmol) and 4-bromobenzaldehyde (4.13 g, 22.3 mmol) in methanol (200 mL) was added 10% Pd/C (1.3 g). The mixture was refluxed overnight, then cooled and filtered though a pad of celite. The filtrate was concentrated under vacuum and purified by chromatography on a silica gel column with 0-10% methanol/dichloromethane to provide the title compound (1.2 g, 17%). MS
(ESI) m/e 317 (M+H)+

2-(4' -formylbipheny-4-yl)-1 H-b enzimidazole-4-carboxamide A solution of EXAMPLE 3A (1.18 g, 3.8 mmol) and 4-formylphenylboronic acid (0.57 g, 3.8 mmol) in dioxane (15 mL) was treated with [1,1'-bis(diphenylphosphino)ferrocene]dichloro palladium (0.32 g, 0.4 mmol) and sodium carbonate (1.8 g, 17 mmol). The mixture was heated at 95 C for 24 hours, then cooled and filtered. The filtrate was concentrated to provide the crude product, which was used without further purification. MS (ESI) m/e 342 (M+H)+.

2- {4'-[(cyclohexylmethylamino)methyl]biphenyl-4-yl} -1H-benzimidazole-4-carboxamide The title compound was prepared as the trifluoroacetate salt using the procedure as described in EXAMPLE IB, substituting EXAMPLE 3B for EXAMPLE 1 A. 'H NMR
(DMSO-d6) S 9.33 (s, 1H), 8.35 (t, J=7.5 Hz, 2H), 7.86-7.93 (m, 2H), 7.78 (s, 3H), 7.73-7.78 (m, 2H), 7.66 (d, J=8.3 Hz, 2H) 7.36 (t, 1=7.82 Hz, 1H), 4.49 (s, 1H), 4.18-4.47 (m, 1H), 3.25 (s, 1H), 2.65 (d, J=4.6 Hz, 3H), 2.10 (m, 2H), 1.85 (s, 2H), 1.61 (s, 1H), 1.47-1.56 (m, 1H), 1.24-1.32 (m, 1H), 1.18 (s, 1H).

- 2-(4'-cyclopropylaminomethylbiphenyl-4-yl)-1H-benzimidazole-4-carboxamide The title compound was prepared as the trifluoroacetate salt using the procedure as described in EXAMPLE 1B, substituting EXAMPLE 3B for EXAMPLE 1 A and cyclopropylamine for cyclohexylmethylamine. 'H NMR (DMSO-D6) 6 9.00 (s, 2H), 8.36 (d, J=8.3 Hz, 2H), 7.95 (d, J=8.3 Hz, 2H), 7.89 (d, J=8.3 Hz, 3H), 7.74 - 7.78 (m, 2H), 7.64 (d, J=8.3 Hz, 3H) 7.37 (t, J=7.7 Hz, 1H), 4.33 (s, 2H), 2.76 (s, 1H), 0.79-0.86 (m, 4H).

2-(4'-cyclobutylaminomethylbiphenyl-4-yl)-1 H-benzirnidazole-4-carboxamide The title compound was prepared as the trifluoroacetate salt using the procedure as described in EXAMPLE 1B, substituting EXAMPLE 3B for EXAMPLE 1A and cyclobutylamine for cyclohexylmethylamine. 'H NMR (DMSO-D6) & 9.05 (s, 2H), 8.36 (d, J=8.3 Hz, 2H), 7.95 (d, J=8.3 Hz, 2H), 7.89 (d, J=8.3 Hz, 3H), 7.73-7.80 (m, 2H), 7.62 (d, J=8.3 Hz, 2H) 7.37 (t, J=7.83 Hz, 1H), 4.11 (s, 2H), 3.66 (s, 1H), 2.20 (m, 5H), 1.80 (m, 2H).

2-[5-(1-cyclopropylaminoethyl)thiophen-2-yl]-1H-benzimidazole-4-carboxamide ' EXAMPLE 6A
5-acetylthiophene-2-carboxylic acid (2-amino-3-carbamoylphenyl) amide To a solution of 5-acetylthiophene-2-carboxylic acid (1.80 g, 10.55 mmol) in pyridine (12 mL) and N,N-dimethylformamide (12 mL) was added 1,1'-carbonyldiimidazole (1.88 g, 11.60 mmol) and the mixture stirred at 45 C for 4 hours. 2,3-Diaminobenzamide dihydrochloride (synthesized as described in US 6737421, column 11, EXAMPLE 2, step (e), 2.36 g, 10.55 mmol) was added and the mixture was stirred at ambient temperature overnight.
The reaction mixture was concentrated and the residue partitioned between ethyl acetate and water. The resulting yellow solid was collected by filtration, washed with water and ethyl acetate and dried to give the title compound (2.91 g, 91%). MS (APCI): 304 (M+1)+.

2-(5-acetylthiophen-2-yl)-1 H-benzimidazole-4-carboxami de To a solution of EXAMPLE 6A (1.70 g) in hot N,N-dimethylformamide (60 mL) was added acetic acid (50 mL) and the solution heated at 125 C for 3 hours. After cooling, the resulting yellow solid was collected by filtration, washed with methanol and dried to give the title compound (1.52 g, 95%). MS (APCI): 286 (M+1)+.

2-[5-(1-cyclopropylaminoethyl)thiophen-2-yl]-1 H-benzimidazole-4-carboxamide EXAMPLE 6B (100 mg, 0.35 nunol) was dissolved in dimethyl sulfoxide (3 mL) with heating. After cooling to ambient temperature, cyclopropylamine (49 L, 0.70 mmol) was added. The solution was stirred at ambient temperature overnight and zinc chloride (48 mg, 0.35 mmol) was added. After an additional 1 hour stirring, sodium cyanoborohydride (44 mg, 0.70 mmol) was added and the mixture stirred at ambient temperature for 3 hours.
Methanol (3 mL) was added and the solution heated at 80 C for 2 days. The reaction mixture was concentrated and the residue suspended in water. Trifluoroacetic acid was added with stirring until a transparent solution formed. This solution was filtered and purified by HPLC (Zorbax, C-18, 250x2.54 column, Mobile phase A: 0.1% trifluoroacetic acid in water;
B: 0.1% trifluoroacetic acid in acetonitrile; 0-100% gradient) to provide the title compound as the trifluoroacetate salt (65.8 mg). 'H NMR (CD3QD): S 0.78-0.84 (m, 1H), 0.86-0.97 (m, 3 H), 1.85 (d, J=7.1 Hz, 3H), 2.69-2.75 (m, 1H), 4.95 (q, J=6.8 Hz, 1H), 7.39 (t, J=7.8 Hz, 1H), 7.44 (d, J=3.7 Hz, 1H), 7.74 (d, J=7.1 Hz, 1H), 7.84 (d, J=4.0 Hz, 1H), 7.95 (d, J=6.8 Hz, 1H).

2- { 5- [ 1-(cyclohexylmethylamino) ethyl] thiophen-2-yl }-1 H-b enzimidazo le-4-carboxam i de The title compound was prepared as the trifluoroacetic acid salt using the procedure as described in EXAMPLE 6C, substituting cyclohexylmethylamine for cyclopropylamine.
'H NMR (CD3OD): S 0.92-1.09 (m, 2H), 1.17-1.37 (m, 3H), 1.64-1.85 (m, lOH), 2.76 (dd, J=12.4, 7.2 Hz, IH), 2.91 (dd, J=12.6, 6.75 Hz, 1H), 4.78-4.86 (m, 1H), 7.37-7.43 (m, 2H), 7.74 (d, J=8.0 Hz, 1H), 7.83 (d, J=3.7 Hz, 1H), 7.95 (d, J=7.7 Hz, 1H).

2-(3'-cyclopropylaminomethylbiphenyl-4-yl)-1 H-benzimidazole-4-carboxamide 2-(3'-formylbipheny-4-yl)-1 H-benzimidazole-4-carboxamide The title compound was prepared using the procedure as described in EXAMPLE
3B, substituting 3-formylphenylboronic acid for 4-formyl-phenylboronic acid. MS
(ESI) m/e 342 (M+H)}.

2-(3'-cyclopropylaminomethylbiphenyl-4-yl)-1 H-b enzimidazole-4-c arboxamide The title compound was prepared using the procedure as described in EXAMPLE
1B, substituting EXAMPLE 8A for EXAMPLE lA and cyclopropylamine for cyclohexylmethylamine. 'H NMR (DMSO-D6) 5 9.38 (d, J=3.0 Hz, 1H), 8.34 (d, J=8.5 Hz, 2H), 7.88-7.93 (m, 3H), 7.74-7.81 (m, 3H), 7.66 (d, J=7.6 Hz, 1H) 7.46 (t, J=7.5 Hz, 1H), 7.35-741 (m, 2H), 3.88 (s, 2H), 3.29 (m, 1H), 0.42 (d, J=4.9 Hz, 2H), 0.36 (s, 2H).

2-(2'-cyclopropylami nomethylbiphenyl-4-yl)-1H-benzimidazole-4-carboxamide 2-(2'-form.ylbipheny-4-yl)-1H-benzimidazole-4-carboxamide The title compound was prepared using the procedure as described in EXAMPLE
3B, substituting 2-formylphenylboronic acid for 4-formyl-phenylboronic acid. MS
(ESI) m/e 342 (M-i-H)+.

2-(2'-cyclopropylaminornethylbiphenyl-4-yl)-1 H-benzimidazole-4-carboxamide The title compound was prepared using the procedure as described in EXAMPLE 2, substituting EXAMPLE 9A for EXAMPLE lA, and cyclopropylamine for cyclobutylamine.
'H NMR (DMSO-d6) S 9.39 (s, 1H), 8.31 (s, 2H), 7.88 (s, 1H), 7.77 (s, 2H), 7.65 (s, 2H), 7.57 (s; 1H), 7.37 (s, 3H), 7.30 (s, 1H), 3.68 (s, 2H), 2.04 (s, 1H), 0.30 (s, 2H), 0.19 (s, 2H).

2-[3-(2-cyclopropylaminoethyl)phenyl]-1 H-benzimidazole-4-carboxamide EXAMPLE l0A
2-(3-[ 1,3 ]dioxolan-2-ylmethylphenyl)-1H-benzimidazol e-4-carboxamide A solution of 3-(1,3-dioxolan-2-ylmethyl)benzoic acid (1.0 g, 4.80 mmol) in a mixture of pyridine (5 mL) and N,N-dimethylformamide (5 mL) was treated with 1,1'-carbonyldiimidazole (0.856 g, 5.28 mmol) at 45 C for 2 hours. 2,3-diaminobenzamide dihydrochloride (synthesized as described in US 6737421, column 11, EXAMPLE 2, step (e), 1.08 g, 4.80 mmol) was added and the mixture stirred at ambient temperature overnight. The mixture was concentrated and the residue purified by flash chromatography on silica gel with 10% methanol in 2:1 ethyl acetate/hexane. The product was dissolved in acetic acid (20 mL) and the solution heated at 60 C for 2 hours and at 80 C for 1.5 hours, cooled and concentrated and the residue partitioned between ethyl acetate and sodium bicarbonate solution. The organic phase was concentrated and the residue purified on silica gel with 10%
methanol in 2:1 ethyl acetate/hexane to give 1.13 g of the title compound. MS
(APCI) m/z 324 (M+H)+.

2-[3-(2-cyclopropylaminoethyl)phenyl]-1 H-benzimidazole-4-carboxamide A solution of EXAMPLE l0A (300 mg, 0.88 mmol) in acetic acid (10 mL) and water (20 mL) was heated at 70 C for 20 hours. After cooling, the solution was concentrated to give a light yellow solid. To a solution of the crude aldehyde (80 mg) in dimethyl sulfoxide (2 mL) and methanol (3 mL) was added cyclopropylamine (60 L) and the mixture stirred at ambient temperature for 1 hour. Sodium cyanoborohydride (54 mg, 0.86 mmol) was added and the mixture heated at 55 C overnight. Volatiles were removed and the residue purified by HPLC (Zorbax, C-18, 250x2.54 column, Mobile phase A: 0.1% trifluoroacetic acid in -water; B: 0.1 % trifluoroacetic acid in acetonitrile; 0-100% gradient) to provide 8.8 mg of the title compound as a trifluoroacetate salt. To a solution of this salt in 1:1 rnethanol/dichloromethane (1 mL) was added 1M HC1 in ether (3 mL).
Concentration afforded the title compound as a hydrochloride salt. 'H NMR (CD3OD): 8 0.91-1.03 (m, 4H), 2.83-2.89 (m, 1H), 3.23-3.29 (m, 2H), 3.49-3.55 (m, 211), 7.70-7.79 (m, 3H), 8.03=8.13 (m, 3 H), 8.20 (s, 1H), 2-(4-cyclopropylaminomethylphenyl)-1 H-b enzimidazole-4-carboxamide 2-(4-diethoxymethylphenyl)- i H-benzimidazole-4-carb oxamide To a suspension of 2,3-diaminobenzamide dihydrochloride (synthesized as described in US 6737421, column 11, EXAMPLE 2, step (e), 5.0 g, 22.3 mmol) in N,N-dimethylacetamide (DMA, 65 mL) was added lithium hydroxide monohydrate (1.87 g, 44.6 mmol) and the mixture heated at 50 C for 20 minutes. After cooling, terephthaldehyde mono(diethylacetal) (5.1 g, 24.48 mmol) in N,N-dimethylacetamide (10 mL) and sodium bisulphite (4.81 g, 40.16 mmol) were added. The mixture was heated at 100 C
for 3 hours, cooled and the solvent removed. The residue was partitioned between ethyl acetate and Water and the organic layer was washed with brine and concentrated: The residue was purified by flash chromatography on silica gel with ethyl acetate to provide the title compound (4.5 g, 54%). MS (DCUNH3) m/z 340 (M+H)+.

2-(4-formylphenyl)-1 H-benzimidazole-4-carboxamide To a suspension of EXAMI'LE 11A (4.5 g, 13.14 mmol) in ethanol (100 mL) and water (100 mL) was added concentrated sulfuric acid (2.9 mL) and the mixture refluxed for 18 hours. The reaction mixture was cooled, partially concentrated, and the residue neutralized with aqueous sodium hydroxide solution. The solid was collected by filtration, washed with water and cold ethanol and dried to provide the title compound (2.5 g, 73%).
MS (DCUNH3) m/z 266 (M+H)+.

2-(4-cyclopropylaminomethylphenyl)-1 H-benzimidazole-4-carboxamide A solution of EXAMPLE 11B (100 mg, 0.38 mrnol) and cyclopropylamine (43 mg, 0.75 mmol) in 1:1 methanol/ N, N-dimethylformamide (5 mL) was stirred at ambient temperature for 2 hours. Sodium cyanoborohydride (47 mg, 0.75 mmol) and zinc chloride (51 mg, 0.38 mmol) were added and the cloudy mixture stirred at ambient temperature for 18 hours. The mixture was concentrated and the residue purified by HPLC (Zorbax C-8, 0.1%
trifluoroacetic acid/acetonitrile/water). The trifluoroacetate salt was dissolved in methanol, treated with anhydrous hydrogen chloride in ether, and concentrated to give the title compound as the hydrochloride salt (41 mg, 35%). 'H NMR (CD3OD): S 0.89-1.03 (m, 4H), 2.77-2.96 (m, 1H), 4.50 (s, 2H), 7.69 (t, J=8.0 Hz, IH), 7.89 (d,.I=8.6 Hz, 2H), 8.01 (d, J=8.3 Hz, 1H), 8.07 (dd, J=7.7, 0.6 Hz, 1H), 8.29 (d, J=8.3 Hz, 2H).

2-(4-cyclobutylaminomethylphenyl)-1 H-benzimidazole-4-carboxamide The title compound was prepared as the hydrochloride salt (60 mg, 50%) using the procedure as described in EXAMPLE 1 1C, and substituting cyclobutylamine for cyclopropylamine. 'H NMR (CD3OD): 5 1.89-2.04 (m, 2H), 2.23-2.46 (m, 4H), 3.83-3.96 (m, 1H), 4.28 (s, 2H), 7.69 (t, J=8.0 Hz, 1H), 7.87 (d, J 8.3 Hz, 2H), 8.01 (d, J=8.3 Hz, 1H), 8.07 (dd, J=7.7, 0.9 Hz, 1 H), 8.29 (d, J=8.6 Hz, 2H).

2-(4-cyclopentylaminomethylphenyl)-1 H-benzimidazole-4-carboxamide The title compound was prepared as the hydrochloride salt (72 mg, 57%) using the procedure as described in EXAMPLE 1 1C, substituting cyclopentylamine for cyclopropylamine. 1H NMR (CD3OD): S 1.64-1.83 (m, 4H), 1.83-1.93 (m, 2H), 2.14-2.30 (m, 2H), 3.63-3.76 (m, 1H), 4.39 (s, 2H), 7.69 (t, J=8.0 Hz, 1H), 7.91 (d, J=8.6 Hz, 2H), 8.01 (dd, J-8.3, 0.9 Hz, 1 H), 8.07 (dd, J=7.7, 0.9 Hz, 1 H), 8.29 (d, .I--8.6 Hz, 2H).

6-chloro-2- {4-[(1,2,3,4-tetrahydronaphthalen-l-ylamino)methyl]phenyl} -1 H-benzimidazole-4-carboxamide 2-amino-5-chloro-3 -nitrob enzamide Step A
Preparation of 2-amino-3-nitrobenzamide To a solution of 2-amino-3-nitrobenzoic acid (prepared using the procedure as described in US6737421, EXAMPLE 2, part b) in dimethoxyethane (DME, 7.1 mL/g), was added thionyl chloride (1.33 equivalents). The mixture was stirred at 50 C
for 12 hours, cooled and slowly added to concentrated ammonium hydroxide (22 equivalents).
The mixture was stirred at 50 C for 2 hours, water was added, and the mixture was cooled and filtered.
The solid was washed with water and isopropanol, and dried under vacuum to give the title compound (89% yield).

Step Preparation of 2-amino-5-chloro-3-nitrobenzamide A solution of the product of step B (5.0 g, 27.6 mmol) in acetonitrile (1250 mL) was treated with N-chlorosuccinimide (3.87 g, 29 mmol) at 60 C for 24 hours.
After copling, the solid was collected by filtration, washed with acetonitrile and dried to give the title compound (4.0 g, 67%). MS (DCUNH3) m/z 216 (M+H)+

2,3 -diamino-5 -chloro-b enzamide To a solution of EXAMPLE 14A (4.0 g, 18.6 mmol) in tetrahydrofuran (500 mL) and ethanol (500 mL) was added Raney nickel (50% in water, 2.0 g). The mixture was stirred under hydrogen (balloon) at ambient temperature for 6 hours. Solid material was filtered off and the filtrate was concentrated to give the title compound (100% yield). MS
(DCI/NH3) m/z 186 (M+H)+.

6-chloro-2-(4-diethoxymethylphenyl)-1 H-benzirnidazole-4-carboxamide The title compound was prepared using the procedure as described in EXAMPLE
11A, substituting EXAMPLE 14B for 2,3-diaminobenzamide dihydrochloride (47%
yield).
MS (DCTINH3) m/z 374 (M+H)'.

6-chloro-2-(4-formylphenyl)-1 H-benzirnidazole-4-carboxamide The title compound was prepared using the procedure as described in EXAMPLE

1 1B, substituting EXAMPLE 14C for EXAMPLE 1 lA (82% yield). MS (DCT/NH3) m/z (n'1-i-H)+=

6-chloro-2- {4-[(1,2,3,4-tetrahydronaphthalen-l-ylamino)methyl]phenyl} -1 H-benzimidazole-4-carboxamide The title compound was prepared as the hydrochloride salt (50% yield) using the procedure as described in EXAMPLE 11C, substituting EXAMPLE 14D for EXAMPLE

and 1,2,3,4-tetrahydro- 1 -naphthalene for cyclopropylamine. 'H NMR (CD3OD): S
1.90-2.00 (m, 1H), 2.00-2.10 (m, 1H), 2.18-2.29 (m, 1H), 2.33-2.44 (m, 1H), 2.83-2.93 (m, 1H), 2.95-3.05 (m, 1H), 4.45-4.55 (m, 2H), 4.65 (t, J=5.0 Hz, 1H), 7.26-7.30 (m, 1H), 7.32 (d, J=7.6 Hz, 1H), 7.35-7.39 (m, 111), 7.50 (d, J=7.6 Hz, 1H), 7.93 (d, J=8.2 Hz, 2H), 8.06 (d, J=1.5 Hz, 1H), 8.13 (d, J=1.8 Hz, 1H), 8.27 (d, J=8.2 Hz, 2H).

2-(4-cyclopropylaminomethylphenyl)-6-fluoro-1 H-benzimidazole-4-carboxamide 2-bromo-4-fluoro-6-nitrophenylamine To a solution of 4-fluoro-2-nitroaniline (20 g, 128 mmol) in dichloromethane (600 mL) and acetic acid (200 mL) was slowly added bromine (13 mL, 256 mmol) at 0 C. The mixture was stirred at 0 C for 1 hour and at ambient temperature for 16 hours.
The mixture was concentrated and the residue partitioned between ethyl acetate and sodium bicarbonate solution. The organic phase was washed with sodium bisulphite solution and concentrated and the residue recrystallized from hexane/dichloromethane to provide the title compound (22.7 g, 76%). MS (DCI/NF-I3) m/z 236 (M+H)+.

2-amino-5-fluoro-3-nitrobenzonitrile A suspension of EXAMPLE 15A (22.7 g, 96.2 mmol), zinc cyanide (22.6 g, 192 mmol) and palladium tetrakis(triphenylphosphine) (7.78 g, 6.7 mmol) in N,N-dimethylformamide (300 mL) was heated at 80 C for 22 hours. After cooling, the mixture was partitioned between ethyl acetate and brine and the organic phase washed with water and concentrated. Recrystallization from methanol provided the title compound (13.9 g, 80%).
MS (DCI/NH3) m/z 182 (M+H)+.

2-amino-5-fluoro-3 -nitro-benzamide A suspension of EXAMPLE 15B (13.9 g, 77 mmol) in polyphosphoric acid (400 g) was stirred at 115 C for 3 hours. After cooling, water and dichloromethane were added and the mixture stirred at ambient temperature for 30 minutes. The solid was filtered and recrystallized from methanol to give the title compound (11.2 g, 74%). MS
(DCUNH3) m/z 200 (M+H)+.

2,3-diamino-5-fluorobenzamide To a solution of EXAMPLE 15C (11.2 g, 56.28 mmol) in tetrahydrofuran (50 mL) and ethanol (50 mL) was added Raney nickel (50% in water, 11.0 g) and the mixture stirred at ambient temperature under hydrogen (60 psi) for 2 hours. The solid material was filtered off and the filtrate concentrated to provide the title compound (9.1 g, 96%).
MS (DCI/NH3) m/z 170 (M+H)+.

EX.AMPLE 15E
2-(4-diethoxymethylphenyl)-6-fluoro-1 H-benzimidazole-4-carboxamide The title compound was prepared using the procedure as described in EXAMPLE
1 1A, substituting EXAMPLE 15D for 2,3-diaminobenzamide dihydrochloride (50%
yield).
MS (DCI/NH3) m/z 358 (M+H)+.

6-fluoro-2-(4-formylphenyl)-1 H-b enzimidazole-4-carboxamide The title compound was prepared using the procedure as described in EXAMPLE
11B, substituting EXAMPLE 15E for EXAMPLE 11A (95% yield). MS (DCI/NH3) m/z (M+H)+.

2-(4-cyclopropylaminomethylphenyl)-6-fluoro-1 H-benzimidazole-4-carbox amide The title compound was prepared as the hydrochloride salt (14% yield) using the procedure as described in EXAMPLE 11 C, substituting EXAMPLE 15F for EXAMPLE
11 B.
'H NMR (CD3OD): S 0.91-1.02 (m, 4H), 2.81-2.92 (m, 1H), 4.49 (s, 2H), 7.72 (dd, .I=7.6, 2.14 Hz, 1H), 7.86 (d, J=8.2 Hz, 2H), 7.88 (d, J=2.1 Hz, 1H), 8.27 (d, J=8.5 Hz, 2H).

2-(4-Cyclobutylaminomethylphenyl)-6-fluoro-1 H-b enzimidazole-4-carboxamide The title compound was prepared as the hydrochloride salt (22% yield) using the procedure as described in EXAMPLE 11C, substituting EXAMPLE 15F for EXAMPLE

and cyclobutylamine for cyclopropylamine. 'H NMR (CD3OD): 6 1.89-2.01 (m, 2H), 2.23-2.33 (m, 2H), 2.33-2.44 (m, 2H), 3.83-3.94 (m, 1H), 4.27 (s, 2H), 7.74 (dd, J-7.6, 2.1 Hz, 1H), 7.8S (d, J=8.2 Hz, 2H), 7.88 (dd, J=9.9, 2.3 Hz, 1H), 8.27 (d, J=8.2 Hz, 2H).

2-(4-Cyclopentylaminomethylphenyl)-6-fluoro-1 H-berizimidazole-4-carboxamide The title compound was prepared as the hydrochloride salt (23% yield) using the procedure as described in EXAMPLE 11C, substituting EXAMPLE 15F forEXANIl'LE

and cyclopentylamine for cyclopropylamine. I H NMR (CD3OD): S 1.66 - 1.82 (m, 4H), 1.82-1.93 (m, 2H), 2.15-2.28 (m, 2H), 3.65-3.69 (m, 1H), 4.39 (s, 2H), 7.75 (dd, J=7.6, 2.1 Hz, 1H), 7.88 (d, J=8.2 Hz, 2H), 7.90 (d, J=2.4 Hz, 1H), 8.27 (d, J=8.5 Hz, 2H).

2-[4-(2-cyclopropylaminoethyl)phenyl]-1 H-benzimidazole-4-carboxamide The title compound was prepared as the hydrochloride salt using the procedure as described in EXAMPLE 10, substituting 4-(1,3-dioxolan-2-ylmethyl)benzoic acid for 3-(1,3-dioxolan-2-ylmethyl)benzoic acid in EXAMPLE 10A. 'H NMR (CD3OD): 8 0.92-1.01 (m, 4H), 2.81-2.88 (m, 1H), 3.21-3.25 (m, 2H), 3.46-3.51 (m, 2H), 7.68 - 7.73 (m, 3 H), 8.02 (d, J=7.6 Hz, 1H), 8.06 (d, J=7.6 Hz, 1H), 8.20 (d, J=7.9 Hz, 2H).

2-[4-(2-cyclobutylaminoethyl)phenyl]-1H-benzimidazole-4-carboxamide The title compound was prepared as the hydrochloride salt using the procedure as described in EXAMPLE 10, substituting 4-(1,3-dioxolan-2-ylmethyl)benzoic acid for 3-(1,3-dioxolan-2-ylmethyl)benzoic acid in EXAMPLE l0A and substituting cyclobutylamine for cyclopropylamine in EXAMPLE l OB. 'H NMR (CD3OD): 8 1.89-2.00 (m, 2H), 2.24-2.32 (m, 2H), 2.33-2.40 (m, 2H), 3.15-3.21 (m, 2H), 3.22-3.28 (m, 2H), 3.81-3.88 (m, 1H), 7.68-7.73 (m, 3H), 8.02 (d, J=8.2 Hz, 1H), 8.06 (d, J=7.6 Hz, 1H), 8.19 (d, J=8.2 Hz, 2H).

2-(4-cyclopropylaminomethyl-2-fluorophenyl)-1 H-benzimidaz ole-4-carboxamide methyl 4-cyano-2-fluorobenzoate A solution ofinethyl 4-bromo-2-fluorobenzoate (10.0 g, 43 mmol), zinc cyanide (10.0 g, 86 mmol) and palladium tetrakis(triphenylphosphine) (2.5 g, 0.64 mmol) in anhydrous N,N-dimethylformamide (100 mL) was purged with nitrogen and the mixture stirred at 80 C
overnight. After cooling, the mixture was partitioned between ethyl acetate and brine and the organic phase washed with water and concentrated. The solid was purified on silica gel using 1:5 ethyl acetate/hexane to afford the title compound (6.1 g, 80%). MS (DCI):
m/z 180 (M+H) rnethyl2-fluoro-4-formylbenzoate EXAMPLE 20A (310 mg, 1.73 mmol) was dissolved in 60% aqueous acetic acid (10 mL) with warming. Raney nickel (60 mg) was added and the mixture was stirred under hydrogen at ambient temperature for 18 hours. Solid material was filtered off and the filtrate concentrated. The residue was purified on silica gel using 1:4 ethyl acetate/hexane to give the title compound (220 mg, 70%). MS (DCI): m/z 183 (M+H)}.

methyl 4-[1,3]dioxolan-2-yl-2-fluorobenzoate A solution of EXAMPLE 20B (2.0 g, 11 mmol), 1,2-ethanediol (1.0 g, l 6 mmol) and p-toluenesulfonic acid monohydrate (10 mg) in benzene (10 mL) was heated under reflux with a Dean-Stark apparatus for about 6 hours. Afler cooling, the mixture was partitioned between ethyl acetate and brine and the organic phase washed with 10% sodium hydroxide solution and water. After concentration, the residue was purified by flash chromatography using 1:5 ethyl acetate/hexane to give the title compound (2.1 g, 80%). MS
(DCI): m/z 227 (M+H)+.

4-[1,3]dioxolan-2-yl-2-fluorobenzoic acid To a solution of EXAMPLE 20C (2.0 g) in tetrahydrofuran (10 mL) and water (5 mL) was added lithium hydroxide monohydrate (1 g) in water (5 mL). Methanol was added until a transparent solution formed and the solution stirred at ambient temperature for 4 hours and concentrated to about 5 mL. The residue was acidified with 2N HCl to pH 2 and the mixture partitioned between ethyl aoetate and water. The organic phase was washed with water, dried over magnesium sulfate, filtered and concentrated to give the title compound (1.5 g, 79%).
MS (DCI): m/z 213 (M+H)+.

2-(4-[1,3] dioxolan-2-yl-2-fluorophenyl)-1H-benzimidazole-4-carboxamide A solution of EXAMPLE 20D (1.5 g, 7.1 mmol) in pyridine (5 mL) and N,N-dimethylformamide (20 mL) was treated with 1,1'-carbonyldiimidazole (1.4 g, 8.5 mmol) at 40 C for 30 minutes. 2,3-Diaminobenzamide dihydrochloride (synthesized as described in US 6737421, column 11, EXAMPLE 2, step (e), 1.58 g, 7.1 mmol) was added and the mixture stirred at ambient temperature overnight. Solvents were removed and the residue suspended in 10 mL of acetic acid. The mixture was stirred at 80 C overnight, cooled, and concentrated and the residue was purified by flash chromatography using ethyl acetate to give the title compound (500 mg, 22%). MS (DCI/NH3) m/z 329 (M+H)}.

2-(2-fluoro-4-formylphenyl)-1 H-benzimidazole-4-carboxamide A solution of EXAMPLE 20E (500 mg, 1.5 mmol) in acetic acid (5 mL) and water (10 mL) was heated at 80 C overnight. After cooling, the mixture was concentrated to give the title compound (400 mg, 94%). MS (DCI/NH3) m/z 285 (M+H)+.

2-[4-(cyclopropylamino)-2-fluorophenyl]-1 H-benzimidazole-4-carboxamide To a solution of EXAMPLE 20F (85 mg, 0.3 mmol) in N,N-dimethylformamide (5 mL) and methanol (10 mL) was added cyclopropylamine (34 mg, 0.6 mmol) and the solution stirred at ambient temperature for 10 minutes. Sodium cyanoborohydride (38 mg) was added and the mixture heated at 55 C overnight. After cooling, the mixture was concentrated and the residue purified by HPLC (Zorbax, C-18, 250x2.54 column, Mobile phase A:
0.1%
trifluoroacetic acid in H20; B: 0.1% trifluoroacetic acid in acetonitrile; 0-100% gradient) to provide the title compound as the trifluoroacetate salt (62 mg). 'H NMR
(CD3OD) S 0.96 (d, J=5.5 Hz, 4H); 2.84 (t, J=5.5 Hz, 1H); 4.42 (s, 2H); 7.43 (t, J=7.8 Hz, 1H);
7.47-7.61 (m, 2H); 7.83 (d, J=8.0 Hz, 1H); 7.97 (d, .I 7.7 Hz, 1H); 8.38 (t, J=7.8 Hz, 1H).

2-[4-(I -cyclopropylaminoethyl)phenyl]-1H-benzimidazole-4-carboxamide A solution of EXAMPLE 1 A (0.07 g, 0.3 mmol) and cyclopropylamine (0.33 mL, 4.8 mmol) in methanol (1 mL) was treated with sodium cyanoborohydride (0.0 16 g, 0.3 mmol) and acetic acid (0.2 mL). The mixture was stirred at arnbient temperature for 48 hours and concentrated. The residue was purified by chromatography on silica gel column with 0-10%

dichloromethane/methanol/0.1% ammonium hydroxide to provide the title compound. 'H
NMR (DMSO-d6) S 9.12 (s, 2H), 8.33 (d, J=8.3 Hz, 2H), 7.88 (d, J=7.4 Hz, IH), 7.71-7.78 (m, 4H), 7.37 (t, J=7.8 Hz, IH), 4.59 (m, 1H), 2.57 (s, 1Hz), 1.64 (d, J=6.6 Hz, 3H), 0.81-0.87 (m, 1H), 0.75-0.79 (m, 1H), 0.66-0.72 (m, 2H).

Example 22 2-(4-cyclobutylaminomethyl-2-fluorophenyl.]-1 H-benzimidazole-4-carboxamide Example 22A
methyl 4-[ 1,3] dioxan-2-yl-2-fluorobenzoate A solution of 2.0 g (11 mmol) of EXAMPLE 20B, 1.67 g (22 mmol) of 1,3-propanediol, 62 mg (1.1 mmol) of 1,2-ethanediol, 20 mL of benzene and 10 mg of p-toluenesulfonic acid monohydrate was heated at reflux for about 6 hours with a Dean-Stark apparatus until water no longer separated. After cooling, the mixture was partitioned between ethyl acetate and brine and the organic phase washed with 10% sodium hydroxide and water and concentrated.
The residue was purified by flash chromatography on silica gel using 1:5 ethyl acetate/hexane) to give the title compound (1.9 g, 73%). MS (DCI): m/z 241 (M+H)".

Example 22B
4-[1,3]dioxan-2-yl-2-fluorobenzoic acid To a solution of EXAMPLE 22A (1.9 g) in tetrahydrofuran (10 mL) and water (5 mL) was added lithium hydroxide (1 g) in water (5 mL). Methanol was added until homogeneous and the solution stirred at ambient temperature for 4 hours. The mixture was concentrated to 5 mL and the residue acidified to pH 2 with 2 N hydrochloric acid. Ethyl acetate and water were added and the organic phase separated, washed with water, dried over magnesium sulfate, filtered and concentrated to give the title compound (1.7g, 95%). MS
(DCI): m/z 227 (M+H)+.

Example 22C
2-(4-[ 1,3 ] dioxan-2-yl-2-fluorophenyl)-1H-benzimidazole-4-carboxamide A solution of EXAMPLE 22B (770 mg, 3.4 rnmol) in pyridine (5 rnL) and N,N'-dimethylformamide (10 mL) was treated with 1,1'-carbonyldiimidazole (763 mg, 3.4 mmol) at 40 C for 30 minutes. 2,3-Diaminobenzamide dihydrochloride (826 mg, 5.1 mmol) was added and the mixttire stirred at ambient temperature overnight. The mixture was concentrated and the residue refluxed overnight in 20 mL of xylene and 2 mL of acetic acid.

After cooling and concentration, the residue was purified by flash chromatography on silica gel using ethyl acetate to give the title compound (550 mg, 50%). MS (DCI/NH3) m/z 342 (M+H)+=

Example 22D
2-(2-fluoro-4-formylphenyl)-1H-benzimidazole-4-carboxamide A solution of EXAMPLE 22C (550 mg, 1.6 mrnol) in acetic acid (5 mL) and water (10 mL) was heated at 70 C overnight. After cooling, the mixture was concentrated to give the title compound (430 mg, 93%). MS (DCUNH3) m/z 285 (M+H)+.

Example 22 2-(4-cyclobutylaminomethyl-2-fluorophenyl)-1 H-benzimidazole-4-carboxamide To a suspension of EXAMPLE 22D (50 mg, 0.17 mmol) in methanol (10 mL) was added cycolobutylamine (36 mg, 0.5 mmol) and the mixture stirred at 50 C for 4 hours. Sodium cyanoborohydride (38 mg) was added and the mixture heated at 55 C overnight.
After cooling, the mixture was concentrated and the residue purified by HPLC
(Zorbax, C-18, 250x2.54 column, Mobile phase A: 0.1 % trifluoroacetic acid in water; B: 0.1 %
trifluoroacetic acid in acetonitrile; 0-100% gradient) to provide the title compound as the trifluoroacetate salt (62 mg). 'H NMR (DMSO-D6): 8 1.70-1.96 (m, 2H); 2.09-2.33 (m, 4H); 3.77 (t, J=7.1 Hz, 1H); 4.18 (s, 2H); 7.40 (t, .1=7.8 Hz, 1H); 7.55 (dd, J 8.0, 1.5 Hz, 1H);
7.65 (dd, J-12.1, 1.4 Hz, 1H); 7.78 (s, 1H); 7.84 (d, J--8.0 Hz, 1H); 7.93 (d, J=8.0 Hz, 1H);
8.38 (t, J=8.0 Hz, 1H); 9.14 (s, 1H) 9.34 (s, 2H).

Example 23 2-(4-cyclohexylasninomethyl-2-fluorophenyl)-1.Fl-benzimidazole-4-carboxamide The title compound was prepared according to the procedure for EXAMPLE 22, substituting cyclohexylamine for cyclobutylamine. 'H NMR (DMSO-D6): 8 1.10-1.19 (m, 1H), 1.20-1.41 (m, 4H), 1.63 (br d, J 12.6 Hz, 1H), 1.80 (br d, J=12.9 Hz, 2H), 2.09-2.17 (m, 2H), 3.03-3.11 (m, 1H), 4.29-4.34 (m, 2H), 7.40 (t, J=8.0 Hz, 1H), 7.57 (dd, J=8.3, 1.5 Hz, 1H), 7.66 (dd, J=12.3, 1.2 Hz, 1H), 7.77 (br s, 1H), 7.82 (d, J=8.0 Hz, 1H), 7.92 (d, J=8.3, 1H), 8.39 (t, J=8.0 Hz, 1H), 8.94 (br s, 2H), 9.14 (br s, 1H).

Example 24 2-(4-cyclopentylaminomethyl-2-fluorophenyl)-1H-benzimidazole-4-carboxamide The title compound was prepared according to the procedure for EXAMPLE 22, substituting cyclopentylamine for cyclobutylamine. 'H NMR (DMSO-d6): S 1.51-1.62 (m, 2H), 1.65-1.78 (m, 4H), 1.96-2.08 (m, 2H), 3.51-3.60 (m, 1H), 4.26-4.33 (m, 2H), 7.41 (t, J=7.7 Hz, IH), 7.59 (d, J=8.0 Hz,.1 H), 7.68 (d, J=12.3 Hz, 1 H), 7.78 (br s, 1 H), 7.84 (d, J=8.0 Hz, 1 H), 7.93 (d, J=7.7 Hz, 1H), 8.38 (t, J=7.8 Hz, 1H), 9.15 (br s, 2H),).

EXAMPLE 25 ( 2- {2-[(cyclopentylamino)methyl]-1,3 -thiazol-4-yl } -1 H-b enzimidazole-4-carboxamide ethyl2-( {[(benzyloxy)carbonyl]amino} methyl)-1,3 -thiazole-4-carboxylate To a solution of N-benzyloxycarbonylglycine thioamide (1.49 g, 6.64 mmol) in 1,2-dimethoxyethane (25 mL) was added potassium hydrogen carbonate (2.66 g, 26.56 mmol) and ethylbromopyruvate (3.47 mL, 27.54 mmol) at -20 C and the mixture stirred at -20 C
overnight. The mixture was filtered through a pad of celite and the filtrate was concentrated and the residue was dissolved in 1,2-dimethoxyethane. After cooling to -20 C, a solution of trifluoroacetic anhydride (2.85 mL, 20.52 mmol) and 2,6-lutidine (5.14 mL, 44.29 mmol) in 1,2-dimethoxyethane (10 mL) was added dropwise over 10 minutes. After stirring for 45 minutes, the solution was concentrated and partitioned between chloroform and water. The organic layer was concentrated and the residue was purified by flash chromatography on silica gel eluting with 60% ethyl acetate in hexanes to provide the title product (1.8 g, 85%):
MS (DCI/NH3) m/z 321 (M+H)+.

2-( {[(benzyloxy)carbonyl]amino}methyl)-1,3-thiazole-4-carboxylic acid To a solution of EXAMPLE 25A (1.8 g, 5.62 mmol) in a 5:1 mixture of methanol and water (120 mL) was added lithium hydroxide monohydrate (1.18 g, 28.09 mmol) and the mixture stirred at ambient temperature for 16 hours. The mixture was partially concentrated and the residue brought to pH 2 using 2M hydrochloric acid. The mixture was partitioned between ethyl acetate and brine and the organic phase was washed with water, dried over magnesium sulfate, filtered and concentrated to provide the title compound (1.23 g, 75%):
MS (DCI/NH3) rn/z 293 (M+H)+.

benzyl {4-[4-(aminocarbonyi)-1H-benzimidazol-2-yl]-1,3-thiazol-2-yl}methylcarbamate A solution of EXAMPLE 25B (1.7 g, 5.82 mmol) in N,N-dimethylformamide (20 mL) and pyridine (20 mL) was treated with 1,1'-carbonyldiimidazole (990 mg, 6.11 mmol) at 50 C for 2 hours. 2,3-Diaminobenzamide dihydrochloride (1.3 g, 5.82 mmol) was added and the mixture stirred at ambient temperature for 16 hours. The mixture was concentrated and the residue was dissolved in acetic acid (50 mL) and heated at 100 C for 2 hours. After cooling, the solution was concentrated and the residue was partitioned between ethyl acetate and saturated sodium bicarbonate. The organic phase was washed with water and concentrated and the residue was purified by flash chromatography on silica gel using 70%
ethyl acetate in hexanes to provide the title compound (1.75 g, 74%): MS
(DCUNH3) mlz 408 (M+H)+.

2-[2-(aminomethyl)-1,3-thiazol-4-yl]-1H-benzimidazole-4-carboxamide A solution of EXAMPLE 25C (1.7 g, 4.17 mmol) in trifluoroacetic acid (15 mL) was heated at 50 C for 16 hours. The mixture was cooled and concentrated and the residue purified by HPLC (Zorbax C-18, 0.1% trifluoroacetic acidlacetonitrile/water) to provide the product as the trifluoroacetate salt. The salt was dissolved in methanol and a solution of 1.8 M hydrochloric acid in diethyl ether (30 mL) added. Concentration afforded the title compound as the hydrochloride salt (910 mg, 80%): MS (DCI/NH3) m/z 274 (M+H)+.

2- {2-[(cyclopentylamino)methyl]-1,3-thiazol-4-yl} -1 H-benzimidazole-4-carboxamide To a solution of EXAMPLE 25D (50 mg, 0.18 mmol) in methanol (5 mL) was added cyclopentanone (32 .L, 0.36 mmol) and triethylamine (25 L). The solution was stirred at ambient temperature for 1 hour and sodium cyanoborohydride (34 mg, 0.54 mmol) was added. After stirring for 60 hours, the mixture was concentrated and the residue purified by HPLC (Zorbax C-18, 0.1% trifluoroacetic acid/acetonitrile/water) to provide the title compound as the trifluoroacetate salt. The salt was dissolved in methanol and a solution of 1.8 M hydrochloric acid in diethyl ether (30 mL) was added. Concentration afforded the title compound as the hydrochloride salt (16 mg, 20%): 'H NMR (CD3OD) 8 1.71-1.77 (m, 2H), 1.79-1.87 (m, 2H), 1.87-1.95 (m, 2H), 2.21-2.30 (m, 2H), 3.79-3.88 (m, 1H), 4.83 (s, 2H), 7.71 (t, J=7.9 Hz, 1H), 8.05 (d, J 8.2 Hz, 1H), 8.09 (d, J=7.0 Hz, 1H), 9.10 (s, IH).

2- {2-[(cyclohexylamino)methyl]-1,3-thiazol-4-yl} -1 H-benzimidazole-4-carboxamide The title compound was prepared as a HCl salt according to procedure described in EXAMPLE 25E, substituting cyclohexanone for cyclopentanone. 'H NMR (CD30D) 8 1.25-1.34 (m, 1H), 1.38-1.48 (m, 2H), 1.48-1.57 (m, 2H), 1.76 (d, J=12.8 Hz, 1H), 1.95 (d, J=13.4 Hz, 2H), 2.28 (d, J=11.9 Hz, 2H), 3.32-3.41 (m, 1H), 4.84 (s, 2H), 7.70 (t, J-7.9 Hz, 1 H), 8.03 (d, J=7.3 Hz, 1 H), 8.08 (d, J=7.6 Hz, 1H), 9.07 (s, 1H).

2- {2-[(cyclobutylamino)methyl]-1,3-thiazol-4-yl} -1 H-benzimidazole-4-carboxamide The title compound was prepared as a HCI salt according to procedure described in EXAMPLE 25E, substituting cyclobutanone for cyclopentanone. Yield: 11%. 'H NMR
(CD3OD) 8 1.93-2.05 (m, 2H), 2.30-2.38 (m, 2H), 2.39-2.47 (m, 2H), 4.00-4.10 (m, 1H), 4.70 (s, 2H), 7.69 (t, J=7.9 Hz, 1H), 8.03 (d, J=8.2 Hz, 1H), 8.08 (d, J--7.6 Hz, 1H), 9.06 (s, 1H).

Claims (17)

1. A compound of Formula (I) or a therapeutically acceptable salt, prodrug, or salt of a prodrug thereof, wherein R1, R2, and R3 are independently selected from the group consisting of hydrogen, alkenyl, alkoxy, alkoxycarbonyl, alkyl, alkynyl, cyano, haloalkoxy, haloalkyl, halogen, hydroxy, hydroxyalkyl, nitro, NR A R B, and (NR A R B)carbonyl;
X1 is selected from the group consisting of aryl and heteroaryl, wherein X1 is optionally substituted with 1, 2, 3, or 4 substituents selected from the group consisting of alkyl, cyano, halogen, and haloalkyl;
W is X2-Y-NR6R7 or Y-NR6R7;
X2 is selected from the group consisting of aryl and heteroaryl, wherein X2 is optionally substituted with 1, 2, 3, or 4 substituents selected from the group consisting of alkyl, cyano, halogen, and haloalkyl;
Y is alkylenyl;
R6 is selected from the group consisting of hydrogen, alkenyl, alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, alkyl, alkynyl, aryl, arylalkyl, cycloalkyl, cycloalkylalkyl, heterocycle, heterocyclealkyl, heteroaryl, heteroarylalkyl, hydroxyalkyl, (NR C R D)alkyl, (NR C R D)carbonyl, (NR C R D)carbonylalkyl, and (NR C R
D)sulfonyl;
R7 is selected from the group consisting of cycloalkyl and cycloalkyl fused to phenyl, wherein R7 may be optionally substituted with 1, 2, or 3 substituents selected from the group consisting of alkenyl, alkoxy, alkoxyalkyl, alkoxycarbonyl, alkoxycarbonylalkyl, alkyl, alkylcarbonyl, alkylcarbonylalkyl, alkynyl, carboxy, cycloalkyl, cycloalkylalkyl, cyano, haloalkoxy, haloalkyl, halogen, hydroxy, hydroxyalkyl, NR C R D, (NR C R
D)alkyl, (NR C R D)carbonyl, (NR C R D)carbonylalkyl, (NR C R D)sulfonyl, and oxo; and R A, R B, R C, and R D are independently selected from the group consisting of hydrogen, alkyl, and alkycarbonyl.
2. A compound of Formula (I) or a therapeutically acceptable salt, prodrug, or salt of a prodrug thereof, wherein R1, R2, and R3 are independently selected from the group consisting of hydrogen and halogen;
X1 is selected from the group consisting of aryl and heteroaryl, wherein X1 is optionally substituted with halogen;

W is X2-Y-NR6R7 or Y-NR6R7;
X2 is aryl;
Y is alkylenyl;
R6 is selected from the group consisting of hydrogen and alkyl; and R7 is selected from the group consisting of cycloalkyl and cycloalkyl fused to phenyl.
3. A compound according to claim 1, wherein R1, R2, and R3 are hydrogen.
4. A compound according to claim 1, wherein W is Y-NR6R7
5. A compound according to claim 1, wherein R6 is hydrogen; and R7 is cycloalkyl.
6. A compound according to claim 1, wherein X1 is thiophenyl.
7. A compound according to claim 1, wherein X1 is thiazolyl.
8. A compound according to claim 1, wherein X1 is phenyl.
9. A compound according to claim 1, wherein X1 is phenyl and X2 is phenyl.
10. A compound according to claim 1 wherein R2 is halogen.
11. A compound or a therapeutically acceptable salt, prodrug, or salt of a prodrug thereof, selected from the group consisting of 2-{4-[1-(cyclohexylmethylamino)ethyl]phenyl}-1H-benzimidazole-4-carboxamide;
2-[4-(1-cyclobutylaminoethyl)phenyl]-1H-benzimidazole-4-carboxamide;
2-{4'-[(cyclohexylmethylamino)methyl]biphenyl-4-yl}-1H-benzimidazole-4-carboxamide;
2-(4'-cyclopropylaminomethylbiphenyl-4-yl)-1H-benzimidazole-4-carboxamide;
2-(4'-cyclobutylaminomethylbiphenyl-4-yl)-1H-benzimidazole-4-carboxamide;
2-[5-(1-cyclopropylaminoethyl)thiophen-2-yl]-1H-benzimidazole-4-carboxamide;
2-{5-[1-(cyclohexylmethylamino)ethyl]thiophen-2-yl}-1H-benzimidazole-4-carboxamide;
2-(3'-cyclopropylaminomethylbiphenyl-4-yl)-1H-benzimidazole-4-carboxamide;
2-(2'-cyclopropylaminomethylbiphenyl-4-yl)-1H-benzimidazole-4-carboxamide;
2-[3-(2-cyclopropylaminoethyl)phenyl]-1H-benzimidazole-4-carboxamide;
2-(4-cyclopropylaminomethylphenyl)-1H-benzimidazole-4-carboxamide;
2-(4-cyclobutylaminomethylphenyl)-1H-benzimidazole-4-carboxamide;
2-(4-cyclopentylaminomethylphenyl)-1H-benzimidazole-4-carboxamide;
6-chloro-2-{4-[(1,2,3,4-tetrahydronaphthalen-1-ylamino)methyl]phenyl}-1H-benzimidazole-4-carboxamide;
2-(4-cyclopropylaminomethylphenyl)-6-fluoro-1H-benzimidazole-4-carboxamide;
2-(4-Cyclobutylaminomethylphenyl)-6-fluoro-1H-benzimidazole-4-carboxamide;
2-(4-Cyclopentylaminomethylphenyl)-6-fluoro-1H-benzimidazole-4-carboxamide;
2-[4-(2-cyclopropylaminoethyl)phenyl]-1H-benzimidazole-4-carboxamide;
2-[4-(2-cyclobutylaminoethyl)phenyl]-1H-benzimidazole-4-carboxamide;
2-(4-cyclopropylaminomethyl-2-fluorophenyl)-1H-benzimidazole-4-carboxamide;
2-[4-(1-cyclopropylaminoethyl)phenyl]-1H-benzimidazole-4-carboxamide;
2-(4-cyclobutylaminomethyl-2-fluorophenyl]-1H-benzimidazole-4-carboxamide;
2-(4-cyclohexylaminomethyl-2-fluorophenyl)-1H-benzimidazole-4-carboxamide;
2-(4-cyclopentylaminomethyl-2-fluorophenyl)-1H-benzimidazole-4-carboxamide;
2-{2-[(cyclopentylamino)methyl]-1,3-thiazol-4-yl}-1H-benzimidazole-4-carboxamide;

2-{2-[(cyclohexylamino)methyl]-1,3-thiazol-4-yl}-1H-benzimidazole-4-carboxamide;
and 2-{2-[(cyclobutylamino)methyl]-1,3-thiazol-4-yl}-1H-benzimidazole-4-carboxamide.
12. A pharmaceutical composition comprising a compound of Formula (I) of claim 1 or a therapeutically acceptable salt thereof, in combination with a therapeutically acceptable carrier.
13. A method of inhibiting poly(ADP-ribose)polymerase (PARP) in a mammal in recognized need of such treatment comprising administering to the mammal a therapeutically acceptable amount of a compound of Formula (I) of claim 1 or a therapeutically acceptable salt thereof.
14. A method of treating inflammation in a mammal in recognized need of such treatment comprising administering to the mammal a therapeutically acceptable amount of a compound of Formula (I) of claim 1 or a therapeutically acceptable salt thereof.
15. A method of treating sepsis in a mammal in recognized need of such treatment comprising administering to the mammal a therapeutically acceptable amount of a compound of Formula (I) of claim 1 or a therapeutically acceptable salt thereof.
16. A method of treating septic shock in a mammal in recognized need of such treatment comprising administering to the mammal a therapeutically acceptable amount of a compound of Formula (I) of claim 1 or a therapeutically acceptable salt thereof.
17. A method of treating cancer in a mammal in recognized need of such treatment comprising administering to the mammal a therapeutically acceptable amount of a compound of Formula (I) of claim 1 or a therapeutically acceptable salt thereof.
CA2628461A 2005-11-15 2006-11-15 Substituted 1h-benzimidazole-4-carboxamides are potent parp inhibitors Expired - Fee Related CA2628461C (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US73692805P 2005-11-15 2005-11-15
US60/736,928 2005-11-15
PCT/US2006/044379 WO2007059230A2 (en) 2005-11-15 2006-11-15 Substituted 1h-benzimidazole-4-carboxamides are potent parp inhibitors

Publications (2)

Publication Number Publication Date
CA2628461A1 true CA2628461A1 (en) 2007-05-24
CA2628461C CA2628461C (en) 2013-09-24

Family

ID=38049280

Family Applications (1)

Application Number Title Priority Date Filing Date
CA2628461A Expired - Fee Related CA2628461C (en) 2005-11-15 2006-11-15 Substituted 1h-benzimidazole-4-carboxamides are potent parp inhibitors

Country Status (10)

Country Link
US (2) US7462724B2 (en)
EP (1) EP1966157B1 (en)
JP (1) JP5162465B2 (en)
CN (1) CN101309908A (en)
AT (1) ATE461923T1 (en)
CA (1) CA2628461C (en)
DE (1) DE602006013191D1 (en)
ES (1) ES2342007T3 (en)
TW (1) TW200736229A (en)
WO (1) WO2007059230A2 (en)

Families Citing this family (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7781596B1 (en) 1998-11-03 2010-08-24 Abbott Laboratories Substituted 2-phenylbenzimidazoles, the production thereof and their use
US7728026B2 (en) * 2005-04-11 2010-06-01 Abbott Laboratories, Inc. 2-substituted-1 h-benzimidazile-4-carboxamides are PARP inhibitors
CN101316834A (en) * 2005-09-29 2008-12-03 艾博特公司 1h-benzimidazole-4-carboxamides substituted with phenyl at the 2-position are potent PARP inhibitors
DE602006013191D1 (en) * 2005-11-15 2010-05-06 Abbott Lab SUBSTITUTED 1H-BENZIMIDAZOLE-4-CARBOXYLIC ACIDS ARE EFFECTIVE PARP INHIBITORS
EP2007733B1 (en) * 2006-04-03 2016-05-25 MSD Italia S.r.l. Amide substituted indazole and benzotriazole derivatives as poly(adp-ribose)polymerase (parp) inhibitors
US20070259937A1 (en) 2006-05-02 2007-11-08 Giranda Vincent L Substituted 1h-benzimidazole-4-carboxamides are potent parp inhibitors
CN101490037B (en) 2006-07-11 2013-06-12 大熊制药株式会社 Biaryl benzoimidazole derivatives and pharmaceutical composition comprising the same
BRPI0806245B1 (en) * 2007-01-10 2022-01-25 Msd Italia S.R.L. Compounds of formula i and their uses
US8067613B2 (en) * 2007-07-16 2011-11-29 Abbott Laboratories Benzimidazole poly(ADP ribose)polymerase inhibitors
US20090062268A1 (en) * 2007-08-27 2009-03-05 Lead Therapeutics, Inc. Novel inhibitors of poly(adp-ribose)polymerase (parp)
CN101970394B (en) * 2007-11-23 2015-05-27 利奥制药有限公司 Novel cyclic hydrocarbon compounds for the treatment of diseases
CN106008460B (en) * 2008-01-08 2022-08-12 默沙东公司 Pharmaceutically acceptable salts of 2- {4- [ (3S) -piperidin-3-yl ] phenyl } -2H-indazole-7-carboxamide
KR101089713B1 (en) 2008-08-20 2011-12-07 한국화학연구원 2-2-Oxo-2-phenylethylsulfanyl-benzimidazole-4-carboxamide derivatives or pharmaceutically acceptable salts thereof, preparation method thereof and pharmaceutical composition containing the same as an active ingredient for the prevention and treatment of the diseases induced by the overactivation of PolyADP-ribosepolymerase-1
CN101619058A (en) * 2009-01-08 2010-01-06 上海交通大学 Benzimidazole-4-acid amide type derivant
WO2010083199A1 (en) * 2009-01-19 2010-07-22 Abbott Laboratories Benzthiazole inhibitors of poly(adp-ribose)polymerase
SG10201401169QA (en) * 2009-04-02 2014-05-29 Merck Serono Sa Dihydroorotate dehydrogenase inhibitors
HUP1000243A2 (en) 2010-05-06 2012-01-30 Avidin Kft 8-hidroxy-quinoline derivatives
WO2013014038A1 (en) * 2011-07-26 2013-01-31 Nerviano Medical Sciences S.R.L. 3-oxo-2,3-dihydro-1h-indazole-4-carboxamide derivatives as parp-1 inhibitors
CN103242273B (en) * 2012-02-09 2015-06-03 中国科学院上海药物研究所 2-arylbenzofuran-7-methanamide compound, preparation method and application thereof
CN104918912B (en) 2012-10-15 2017-02-22 阿尔比马尔公司 Processes for the synthesis of 2-amino-4,6-dimethoxybenzamide and other benzamide compounds
CN102964354B (en) * 2012-11-16 2014-08-13 江苏先声药业有限公司 Thienoimidazole-like derivatives and applications thereof
EP3265560B1 (en) 2015-03-02 2021-12-08 Sinai Health System Homologous recombination factors

Family Cites Families (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB1354554A (en) 1970-08-31 1974-06-05 Robins Co Inc A H 2-substituted benzimidazoles
BE789679A (en) * 1971-10-05 1973-04-04 Leo Pharm Prod Ltd NEW DERIVATIVES OF 5-AMINO ISOPHTHALIC ACID AND THEIR PREPARATION PROCEDURES
US4093726A (en) * 1976-12-02 1978-06-06 Abbott Laboratories N-(2-benzimidazolyl)-piperazines
DE3522230A1 (en) 1985-06-21 1987-01-02 Thomae Gmbh Dr K NEW 2-ARYLIMIDAZOLES, MEDICINAL PRODUCTS CONTAINING THESE COMPOUNDS, AND METHOD FOR THE PRODUCTION THEREOF
DE3830060A1 (en) 1988-09-03 1990-03-15 Boehringer Mannheim Gmbh 2-Phenylbenzimidazoles - process for their preparation and medicaments containing these compounds
CA2225465A1 (en) 1995-08-02 1997-02-13 Newcastle University Ventures Limited Benzimidazole compounds
JP2001515482A (en) 1997-03-07 2001-09-18 メタバシス・セラピューティクス・インコーポレイテッド Novel benzimidazole inhibitors of fructose-1,6-bisphosphatase
US6911462B2 (en) * 1998-05-22 2005-06-28 Avanir Pharmaceuticals Benzimidazole compounds for regulating IgE
GB9815880D0 (en) * 1998-07-21 1998-09-16 Pfizer Ltd Heterocycles
US7781596B1 (en) 1998-11-03 2010-08-24 Abbott Laboratories Substituted 2-phenylbenzimidazoles, the production thereof and their use
CA2350734C (en) 1998-11-17 2006-10-10 Basf Aktiengesellschaft 2-phenylbenzimidazoles and 2-phenylindoles, and production and use thereof
DE19916460B4 (en) 1999-04-12 2006-12-21 Abbott Gmbh & Co. Kg Substituted benzimidazoles, their preparation and use
USRE39608E1 (en) * 1998-11-27 2007-05-01 Abbott Gmbh & Co. Kg Substituted benzimidazoles and their use as PARP inhibitors
DE19918211A1 (en) * 1999-04-22 2000-10-26 Basf Ag New 2-carbocyclyl-benzimidazole-carboxamide derivatives, are PARP inhibitors useful e.g. for treating neurodegenerative disease, epilepsy, ischemia, tumors, inflammation or diabetes
DE19920936A1 (en) * 1999-05-07 2000-11-09 Basf Ag Heterocyclically substituted benzimidazoles, their preparation and use
AU7314200A (en) 1999-09-17 2001-04-24 Yamanouchi Pharmaceutical Co., Ltd. Benzimidazole derivatives
WO2001021634A1 (en) 1999-09-21 2001-03-29 Lion Bioscience Ag Benzimidazole derivatives and combinatorial libraries thereof
DE10021468A1 (en) 2000-05-04 2001-11-08 Basf Ag Use of PARP inhibitors in cosmetic preparations
JP4734705B2 (en) 2000-10-31 2011-07-27 三菱化学株式会社 Positive electrode material for lithium secondary battery, positive electrode for lithium secondary battery, and lithium secondary battery
WO2002068407A1 (en) 2001-02-28 2002-09-06 Yamanouchi Pharmaceutical Co., Ltd. Benzimidazole compound
AU2002334355A1 (en) 2001-09-06 2003-03-18 Prochon Biotech Ltd. Protein tyrosine kinase inhibitors
AU2003230401A1 (en) 2002-05-13 2003-11-11 Icagen, Inc. Bis-benzimidazoles and related compounds as potassium channel modulators
US20040034078A1 (en) 2002-06-14 2004-02-19 Agouron Pharmaceuticals, Inc. Benzimidazole inhibitors of poly(ADP-ribosyl) polymerase
AU2003297925A1 (en) 2002-12-13 2004-07-09 Smithkline Beecham Corporation Thrombopoietin mimetics
KR100791252B1 (en) 2003-01-23 2008-01-03 크리스탈지노믹스(주) Glycogen synthase kinase 3beta inhibitor, composition and process for the preparation thereof
HU0301154D0 (en) 2003-04-28 2003-07-28 Hideg Kalman Dr Pharmaceutical composition
EP1620413A2 (en) 2003-04-30 2006-02-01 Cytokinetics, Inc. Compounds, compositions, and methods
US7728026B2 (en) * 2005-04-11 2010-06-01 Abbott Laboratories, Inc. 2-substituted-1 h-benzimidazile-4-carboxamides are PARP inhibitors
TWI375673B (en) * 2005-04-11 2012-11-01 Abbott Lab 1h-benzimidazole-4-carboxamides substituted with a quaternary carbon at the 2-position are potent parp inhibitors
CN101316834A (en) * 2005-09-29 2008-12-03 艾博特公司 1h-benzimidazole-4-carboxamides substituted with phenyl at the 2-position are potent PARP inhibitors
DE602006013191D1 (en) * 2005-11-15 2010-05-06 Abbott Lab SUBSTITUTED 1H-BENZIMIDAZOLE-4-CARBOXYLIC ACIDS ARE EFFECTIVE PARP INHIBITORS
EP2329818A1 (en) * 2006-01-17 2011-06-08 Abbott Laboratories Combination therapy with PARP inhibitors
US20070259937A1 (en) * 2006-05-02 2007-11-08 Giranda Vincent L Substituted 1h-benzimidazole-4-carboxamides are potent parp inhibitors
US8067613B2 (en) * 2007-07-16 2011-11-29 Abbott Laboratories Benzimidazole poly(ADP ribose)polymerase inhibitors

Also Published As

Publication number Publication date
CN101309908A (en) 2008-11-19
WO2007059230A2 (en) 2007-05-24
ES2342007T3 (en) 2010-06-30
JP2009515986A (en) 2009-04-16
US20070112047A1 (en) 2007-05-17
EP1966157A2 (en) 2008-09-10
US7595406B2 (en) 2009-09-29
WO2007059230A3 (en) 2007-08-02
US20070270476A1 (en) 2007-11-22
US7462724B2 (en) 2008-12-09
DE602006013191D1 (en) 2010-05-06
CA2628461C (en) 2013-09-24
EP1966157B1 (en) 2010-03-24
ATE461923T1 (en) 2010-04-15
JP5162465B2 (en) 2013-03-13
TW200736229A (en) 2007-10-01

Similar Documents

Publication Publication Date Title
US7462724B2 (en) Substituted 1H-benzimidazole-4-carboxamides are potent PARP inhibitors
CA2623822C (en) 1h-benzimidazole-4-carboxamides substituted with phenyl at the 2-position are potent parp inhibitors
JP5228237B2 (en) Substituted 1H-benzimidazole-4-carboxamide is a potent PARP inhibitor
EP2069351B1 (en) Pyrazoloquinazolinones as parp inhibitors
US8217070B2 (en) 2-substituted-1H-benzimidazole-4-carboxamides are PARP inhibitors
JP4960342B2 (en) 1H-benzimidazole-4-carboxamide substituted at position 2 by a quaternary carbon is a potent PARP inhibitor

Legal Events

Date Code Title Description
EEER Examination request
MKLA Lapsed

Effective date: 20171115